{ $- 4’ 23:: . ' ‘ '1‘ $ '3‘ l.- . 4‘ .4 a... . .....p a...” -1 Wu :7}; tab ’:'::T A -.._. .‘.LJ.'« u-.-—. - 4‘21: .- ..'.,-. .1w4—‘ L !‘ \vu PM 2.5!: Jay 4 fig. 0 ‘ 411‘ 1 {”3" $5} f’iem i mg; +3 ’1. {7,1 ‘E" 2.... dazzlp‘ i . I' ‘ “M? §. {'4 5“ (g! 1:2;- 1.1. if?! MM n I ism“; “if ' 3. “'6: :ytawi, gem? rt“ ‘ u ‘ ‘ ‘1}; .I“ '23??? KN; 4: n; I 4.— 11:1. "07- v 3 . 1". flan“? u‘ql,‘ 31‘1”? u-g; fl?" 9333' v '::""» ‘0 ."S .'.' ”333' €5"".’!¢Ms!fl~ 311113 , a; . . :J :, A: ”$311.; 3? 21:“???3 333v! . w: " -: 7:. ‘ "311 ~ h 4: ~ _1 ‘3“: i ’é‘gtz‘I-I‘ We!" 1 ' -' 'i‘ .‘J - .gf'.t - 1'2" gzi'wf 3:25. .‘ :32... j :' n .1 i".- I 2' Wm . ' ":5 '- ‘ 1:53.451: 3;} ,-; ' L . ‘ ' 11““: J - It", ‘ .' ‘. ‘F; t " . a,” 331m ‘ 4’s? .: :3 . . .. > :9 . W “t.“ :1 . ,- .;. hi I .ri. if; A} . z f .‘L‘ '33- I..-‘ x‘ '. YHES‘S RSITY UBRARIES “W M .mmmm Mixing H\ \\| 31293307 m N This is to certify that the dissertation entitled NUCLEAR EXPORT OF A PRE-mRNA SPLICING FACTOR GALECTIN-3 presented by YEOU-GUANG TSAY has been accepted towards fulfillment of the requirements for Eh. 12. degree in WM 1 , Lie—1 U Major professor ) Date X'22'77 .MSU is an Affirmative Action/Equal Opportunity Institution 0- 12771 LIBRARY Michigan State University PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MTE DUE MTE DUE DATE DUE 1/93 campus-p.14 NUCLEAR EXPORT OF A PRE-mRNA SPLICING FACTOR, GALECTIN-3 By Yeou-Guang Tsay A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Biochemistry 1 997 ABSTRACT NUCLEAR EXPORT OF A PRE-mRNA SPLICING FACTOR, GALECTIN-3 By Yeou-Guang Tsay Previous immunofluorescence and subcellular fractionation studies have documented that galectin—3 (Mr ~ 30,000) distributes differentially between the nucleus and the cytoplasm, depending on the proliferative state of the cells under analysis. Thus, it appears that controlled nuclear versus cytoplasmic distribution of galectin-3 may be one of the mechanisms regulating its function. The goal of this thesis was to study parameters that play a role in determining this distribution; one such parameter turned out to be the export of the protein from the nucleus to the cytoplasm. Using digitonin-penneabilized 3T3 cells, we provide evidence that galectin-3 is rapidly and selectively exported from the nucleus. Although both phosphorylated and nonphosphorylated isoforms of galectin-3 are found in the nuclear fiaction, only phosphorylated galectin-3 is identified in the exported fraction, implying that phosphorylation is important forthe nuclear export of the protein. The rate of galectin-3 export is decreased by cold temperature and by the addition of either wheat germ agglutinin or vanny cations (V 0"). More strikingly, galectin-3 export can be inhibited, at least partially, by the simultaneous addition of a peptide bearing a nuclear export signal plus a dinucleotide analog of the cap structure found at the 5’-end of mRNAs. These results suggest that galectin-3 may be exported in association with ribonucleoprotein complexes containing monomethylated cap structure as well as polypeptides containing nuclear export sequences. The transported fraction of the nuclear export assay was analyzed in terms of the polypeptide and RNA components. Gel filtration of the exported nuclear material and analysis for galectin-3 showed that the lectin can be found in at least two sets of high molecular weight complexes (~650 RD and ~60 kD). In the presence of the saccharide ligand, lactose, both of these complexes are disrupted and galectin-3 chromatographs to a position corresponding to ~30 kD polypeptide. The polypeptide components of the high molecular weight complexes containing galectin-3 are specifically revealed by affinity adsorption on a lactose-agarose column, specific elution by lactose, and gel electrophoresis. These polypeptides are not bound to a control cellobiose column. The transported fraction of the nuclear export assay also contains RNA. In the low molecular weight range, the RNA species include tRNA (~80 nucleotides) as well as RNAs of ~100, 300, and 650 nucleotides. High molecular weight RNAs, ranging from ~1 kb to 5 kb, include poly (A)+ mRNA as revealed by hybridization with an oligo(dT) probe. Compounds containing vanadyl cations, which inhibit the export of galectin-3, also inhibit the export of RNAs. All of these results are consistent with the notion that galectin-3 is associated with a ribonucleoprotein complex during its export fiom the nucleus. Copyright by Yeou-Guang Tsay 1 997 To my parents and Huifen ACKNOWLEDGMENTS I would like to express my gratitude to my mentor, Dr. John Wang, who has provided me every available opportunity that can help me to become a better scientist. I enjoyed all of the discussions that we had in these years, which have broadened my view over the field greatly. I would like to thank my guidance committee, Drs. Don Jump, Ron Patterson, El Schindler, and Bill Smith for their criticism and advice, which has been a major part of this work. I thank former and present members of Wang and Schindler’s labs for their friendship and encouragement, especially Dr. Eric Arnoy, Sharon Grabski, Nancy Lin, Mark Kadrofske, Patty Voss and Dr. Anandita Vyakarnam. They have provided generous help to myself as well as my family. Special thanks will be given to Patty and Nancy, who have been helping me to complete my dissertation. Finally, I feel very grateful to my wife. She has accompanied me through the ups and downs in these years. With her love and support, life is much more pleasant and colorful. TABLE OF CONTENTS TABLE OF CONTENTS ....................................................................... vii LIST OF TABLES ................................................................................ ix LIST OF FIGURES ............................................................................... x LIST OF ABBREVIATIONS ................................................................. xiii CHAPTER I: LITERATURE REVIEW ...................................................... 1 INTRODCUTION TO GALECTINS ................................................. 1 INTRODUCTION TO NUCLEAR TRANSPORT ................................ 22 CHAPTER II: NUCLEAR EXPORT OF GLAECTIN-3 IN MOUSE 3T3 FIBROBLASTS: PARAMETERS OF THE TRANSPORT IN DIGITONIN-PERMEABILIZED CELLS ................................. 44 SUMMARY ...................................................... 45 INTRODUCTION ....................................................................... 46 EXPERIMENTAL PROCEDURES ................................................... 50 RESULTS ................................................................................ 58 DISCUSSION ............................................................................ 74 CHAPTER III: NUCLEAR EXPORT OF GALECTIN-3 IN MOUSE 3T3 FIBROBLASTS: CHARACTERIZATION OF THE EXPORTED COMPONENTS ................................................................. 80 SUMMARY ............................................................................... 81 INTRODUCTION ....................................................................... 82 vii EXPERIMENTAL PROCEDURES .................................................. 84 RESULTS ................................................................................ 89 DISCUSSION .......................................................................... 106 CONCLUSIONS .............................................................................. 1 10 LIST OF REFERENCES ..................................................................... 112 viii CHAPTER I 4. LIST OF TABLES SUMMARY OF GALECTINS ................................................ 3 VERTEBRATE NUCLEAR PORE PROTEINS .......................... 25 SIGNALS AND RECEPTORS INVOLVED IN NUCLEAR IMPORT ......................................................................... 29 SIGNALS AND RECEPTORS INVOVLED IN NUCLEAR EXPORT .................................................................................... 38 i) CHAPTER I CHAPTER II 1. LIST OF FIGURES A PHYLOGENETIC TREE FOR CROWN EUKARYOTES ............ 2 INTERACTION OF BOVINE GALECTIN-l WITH N- ACETYLLACTOSAMINE .................................................... 6 SCHEMATIC REPRESENTATION OF THE INVOVLEMENT OF SMALL RIBONUCLEOPROTEIN PARTICLES (snRNP) AND NON- snRNP PROTEINS IN PRE-mRNA SPLICING .......................... 17 ROLES OF GALECTIN CRD IN PRE-mRNA SPLICING ............. 20 SCHEMATIC REPRESENTATION OF A CONCENSUS MODEL OF THE MEMBRANE-BOUND NUCLEAR PORE COMPLEX .......... 23 DIAGRAM SUMMARIZING LOCALIZATIONS OF CHARACTERIZED NUCLEOPORINS WITHIN THE NUCLEAR PORE COMPLEX ............................................................. 27 NUCLEAR IMPORT OF NLS-CONTAININ G PROTEINS HAS TWO DISTINCT STEPS: DOCKING AND TRANSLOCATION ............ 31 (A) A MODEL ILLUSTRATING RAN GTPASE CYCLE. (B) COORDINATION OF PROTEIN IMPORT BY RAN GTPASE CYCLE ........................................................................... 33 SCHEMATIC ILLUSTRATION OF THE DIGITONIN- PERMEABILIZATION CELL SYSTEM FOR THE ASSAY OF NUCLEAR EXPORT ......................................................... 53 COMPARISON OF THE IMMUNOFLUORESCENCE STAINING FOR GALECTIN-3 IN 3T3 CELLS BEFORE AND AFTER DIFFERENTIAL PERMEABILIZATION ................................. 59 CHAPTER III 1. WESTERN BLOTTING ANALYSIS FOR GALECTIN-3 IN THE NUCLEAR FRACTION (NF) AND THE TRANSPORTED FRACTION (TF) BEFORE AND AFTER A 30-MIN EXPORT ASSAY ........................................................................... 61 TEMPERATURE DEPENDENCE OF THE KINETICS OF GALECTIN-3 EXPORT IN THE DIGITONIN PERMEABILIZED CELL ASSAY .................................................................. 63 TWO-DIMENSIONAL GEL ELECTROPHORETIC ANALYSIS FOR GALECTIN-3 IN NF BEFORE THE EXPORT ASSAY (A) AND IN THE TF AFTER AN 8-MIN EXPORT ASSAY (B) ....................... 66 COMPARISON OF SEVERAL NUCLEAR PROTEINS IN TERMS OF THEIR EXPORT OR RETENTION IN THE DIGITONIN- PERMEABILIZED CELL ASSAY ......................................... 68 THE EFFECT OF WHEAT GERM AGGLUTININ (WGA) ON THE EXPORT OF GALECTIN-3 AND RAN IN THE DIGITONIN- PERMEABILIZED CELL ASSAY ......................................... 70 THE EFFECTS OF S’-CAP STRUCTURE NUCLEOTIDE ANALOGS AND OF A PEPTIDE BEARING A NUCLEAR EXPORT SIGNAL (NES) ON THE EXPORT OF GALECTIN-3 AND RAN IN THE DIGITONIN-PERMEABILIZED CELL ASSAY ........................ 72 SCHEMATIC DIAGRAM ILLUSTRATING THE ASSOCIATION OF GALECTIN-3 WITH A RIBONUCLEOPROTEIN COMPLEX IN TF OF THE EXPORT ASSAY ................................................... 73 GELFILTRATION OF THE EXPORTED NUCLEAR COMPONENTS IN TF AND ANALYSIS FOR GALECTIN-l AND -3 ................... 91 THE EFFECT OF LAC ON THE GEL FILTRATION PROFILE OF GALECTIN-l AND GALECTIN-3 IN TF ................................. 93 AFFINITY ADSORPTION OF THE POLYPEPTIDE COMPONENTS IN TF ON LACTOSE AGAROSE AND CELLOBIOSE-AGAROSE ..................................................................................... 95 THE EFFECT OF VANADYL CATIONS (V 02+) ON THE NUCLEAR EXPORT OF GALECTIN-3. ASSAYED BY IMMUNOBLOT'TING NF ................................................................................. 99 THE EFFECT OF VANAYL CATIONS (V 02*) ON THE NUCLEAR EXPORT OF GALECTIN-3 BY IMMUNOFLUORESCENCE ANALYSIS OF NF ........................................................... 100 THE EFFECTS OF VANADYL CATIONS (V 02+) ON THE NUCLEAR EXPORT OF RAN, ASSAYED BY IMMUNOFLUORESCENCE ANALYSIS OF NF ...................... 102 7. GEL ELECTROPHORETIC ANALYSIS OF THE RNA COMPONENTS IN TF WHEN EXPORT ASSAY IS CARRIED OUT IN THE ABSENCE AND PRESENCE OF VANDYL SULFATE. ....103 xii ATP: BR: CBC: CBP: CRD: DNA: ER: FGF: FITC: GDP: GlcNAc: GTP: HIV: hnRNP: Lac: WCK: MIP: mRNA: NEPHGE: LIST OF ABBREVIATIONS adenosine triphosphate Balbiani ring cap binding complex cap binding protein carbohydrate recognition domain deoxyribonucleic acid endoplasmic reticulum fibroblast growth factor fluorescein isothiocyanate guanosine diphosphate N-acetylglucosamine guanosin tn'phosphate human immunodeficiency virus heterogeneous ribonucleoprotein complex lactose Madin-Darby canine kidney M9 interacting protein messenger ribonucleic acid nonequilibrium pH gradient electrophoresis xiii NES: NTF: PAGE: PKI: RanGAP: RIP: SAP: SDS: snRNP: SV40: T-TBS: WGA: nuclear export signal nuclear fraction nuclear localization signal nuclear pore complex nuclear transport factor polyacrylamide gel electrophoresis protein kinase A inhibitor Rev activation domain Ran GTPase activating protein Rev interacting protein ribonucleoprotein spliceosome-associated protein sodium dodecyl sulfate small nuclear ribonucleoprotein simian virus 40 transported buffer transported fraction thimerosol-Tris buffered saline vanadyl ribonucleoside complex wheat germ agglutinin xiv CHAPTER 1 LITERATURE REVIEW INTRODUCTION TO GALECT INS Galectins are a family of proteins defined by their affinity toward galactose/lactose and sequence similarity at the characteristic carbohydrate binding domain (Barondes er al., 1994; Kasai and Hirabayshi, 1996; Leffler, 1997). Galectins were previously found in species in the animal kingdom, including sponges, nematodes, fish, amphibian, birds and mammals. Hence, galectins were considered as a group Of animal lectins for years (Kasai and Hirabayashi, 1996). However, the discovery of a galectin in inky cap mushroom, Coprinus cinereus, challenged this concept (Cooper et al., 1997). This finding not only confirms that galectins can be outside the animal kingdom, but also implies that a common ancestor for animals and fungi may contain galectin-like polypeptides (Figure 1). The evolution of galectins has been an interesting topic. For instance, they have been expanded into a large group of homologs in mammals (Table I), in which at least ten galectins have been identified (Table 1). On the contrary, it is surprising to see that galectins seem to be completely abandoned in some descendants, like yeasts and insects (Cooper et al., 1997) . Plants Prolifera (Sponges) Anthropoda (Insects) Nematoda (Nematodes) Chorda (Birds, Amphibians, Fish, Mammals) Basidiomycota (mushroom) Ascomycota (Yeasts) Figure l. A phylogenetic tree for crown eukaryotes. Galectins have been found in animals, like sponges, nematodes, and vertebrates, as well as a fungus, inky cap mushroom. Table 1. Summary of Galectins Designations MW Structur Tertiary Source Organisms Tissue/Cell Distribution (kDa) :11 Type Structure Mammals Galectin-l 14.5 Proto Dimer Human, rat, mouse, Muscle, heart, lung, hamster, monkey, ox, placenta, brain, spleen, pig liver, lymph node, thymus, prostate, colon Galectin-Z 14.5 Proto Dimer Human, mouse Small intestine Galectin-3 29-35 Chimera Monomer Human, rat, mouse, Macrophage, colon, dog, hamster leukemia cell, fibroblasts Galectin-4 36 Tandem Monomer Human, rat, pig, Alimentary tract epithelium repeat mouse Galectin-S 17-18 Proto Monomer Rat Erythrocyte Galectin-6 34 Tandem Monomer Mouse Castro-intestine repeat Galectin-7 14.5 Proto ? Human, rat Skin Galectin-8 34 Tandem Monomer Human, rat Liver, lung, kidney repeat Galectin-9 35 Tandem ? . Human, rat, mouse Kidney, thymus, Hodgkin's repeat lymphoma Galectin-IO 17 Proto Dimer Human Birds Chick 14K 14 Proto Monomer Chick Skin, intestine, etc. Chick 16K 16 Proto Dimer Chick Muscle, liver, etc. Chick 30K 30 Chimera ? Chick Chondrocyte Amphibians Xenopus 16K 16 Proto Dimer Xenopus Iaevis Skin Bufo 15K 14.5 Proto Dimer B. Arenarum Oocyte Fish Electrolectin l6 Proto Dimer Electric eel Electric organ Congerin l6 Proto Dimer Conger eel Skin mucus Nematodes Nematode 32K 32 Tandem Monomer C. elegans Cuticle, pharynx repeat , Nematode 16K 16 Proto Dimer C. elegans ? OvaGalBP 32 Tandem 7 0. volvulus ? repeat sponges Gth1/2 13-18 Proto Dimer G. cydonium Plasma membrane Fungi Cgl-I/II 15.5/ Proto Dimer C. cinereus Fruiting body 17 4 The galectin structure also went through some changes. Chimera-type galectins are only found in vertebrates but not in primitive animals, implying that they are evolved later than proto- and tandem-repeat type galectins (Table 1). In addition, it seems that activities other than carbohydrate binding has been introduced into the galectin structure. For instance, galectin-10, the Charcot-Leyden crystal protein, was reported to have a lysophospholipase activity (Weller et al., 1982; Ackerman et al., 1993). In the following section, I will focus on the structural characteristics, intracellular sorting, and biological functions of galectins. Since galectin-l and galectin-3 are the most studied galectins, I will discuss properties of the whole family using them as examples. STRUCTURAL CHARACTERISTICS Galectins are categorized into proto-, chimera-, and tandem-repeat types according to their primary structure. According to their molecular architecture, all galectins can be grouped into the following three types: proto-, tandem-repeat and chimera types (Hirabayashi et al., 1992) (Table 1). Prototype, represented by mammalian galectin-1 is composed of only a single carbohydrate recognition domain (CRD) motif. Tandem-repeat type galectins, exemplified by galectin-4, consist of two homologous CRD domains. Chimera-type galectin, represented by galectin-3 is made of a CRD and a nOn-lectin domain. Carbohydrate recognition domain is the signature motif for galectin family. The galectin family has a signature structural motif, carbohydrate recognition domain, which confers saccharide-binding specificity. This domain consists of about 135 amino 5 acids that fold into a B-sandwich with 5- and 6-stranded B-sheets as shown by X-ray diffraction studies on galectin-1 and galectin-2 (Lobsanov et al., 1993; Liao et al., 1994). All of the amino acid residues important for binding with the bound N-acetyllactosamine ligand are found on a continuous stretch of the polypeptide chain, which makes up four antiparallel B-strands and is designated as carbohydrate-binding cassette (Gitt et al., 1992; Lobsanov and Rini, 1997). Galectins forming homodimers, like galectin-l and galectin-2, may contain another structural motif, dimer interface. This motif is formed by two B—strands at the end away from the carbohydrate-binding cassette . Carbohydrate-binding cassette is conserved among galectin family. Amino acid residues that are involved in carbohydrate binding are highly conserved among galectins (Lobsanov et al., 1993; Liao et al., 1994). Analysis of the genomic DNA structure of a number of galectins has also shown that they are all included on a single DNA exon (Barondes et a1. , 1994; Lobsanov and Rini, 1997). There are eight major amino acids in CRDs that directly interact with a disaccharide ligand, like N-acetyllactosamine (Lobsanov et al., 1993; Liao et al., 1994; Ahmed and Vasta, 1994). Among these amino acids, five residues are invariably conserved in all galectins. They primarily interact with the lactose moiety of the ligand (Figure 2). On the other hand, a lot of variations occur at the other three amino acids. They primarily interact with the N-acetylglucosamine moiety of the ligand. Ahmed and Vasta (1994) found that complete identity with galectin-l CRD at the latter three residues is found in a number of the galectins, whereas substitution and deletion occur at these positions in other galectins. Based on identity of these three Figure 2. Interaction of bovine galectin-l with N-acetyllactosamine. Amino acids boxed by lighter shade are completely conserved in all galectins except mammalian galectin-10. Amino acids highlighted by darker shade have greater variations among galectin family. (Modified from Ahmed and Vasta, 1994) 7 amino acids with the galectin-1 CRD, carbohydrate recognition domains of galectins can be classified as two types: type I (conserved) and type II (variable) CRDs (Ahmed and Vasta, 1994). So far, only chicken 14-kDa, chicken 16 kDa, and Bufo 15-kDa galectins belong to the type I category. Type I CRDS have very similar fine carbohydrate-binding specificity, presumably due to the extensive identities at the carbohydrate-binding cassette. On the other hand, type II CRDs have very different carbohydrate recognition properties, reflecting the variations in the cassette motif. The carbohydrate-binding cassette in galectin-10 is unusually varied at these positions, compared with other galectins (Ackerman et al., 1993). Different amino acids are found at the positions that are completely conserved in other galectins. This is perhaps the structural basis for its weaker sugar binding activity. Probably, in order to accommodate a lysophospholipase activity as well as lectin activity, some compromise had to be made on the primary structure. Galectin-l/galectin-2 dimer interface mediates the homodimerization of galectin subunits. The dimer interface for galectin-1 and galectin-2 is mainly made up Of hydrophobic amino acid residues, which form nonpolar surfaces (Lobsanov et al., 1993; Liao et al., 1994). This interface mediates the formation of a 2-fold symmetric anti-parallel dimers seen in galectin-1 and galectin-2. Recent x-ray crystallography of galectin-3 CRD (Kanigsberg et al. 1997) and N-tenninal CRD of galectin-4 (Kayden et al., 1997) have shown that the nonpolar surfaces are essentially eliminated by the presence of hydrophilic residues. Lobsanov and Rini (1997) argued that the disruption of nonpolar surfaces prevents galectin-3 or galectin-4 from forming homodimer complex. However, whether CRD of galectin-3 or galectin-4 forms dimers is still controversial. 8 For instance, when isolated C-domain of galectin-3 was analyzed by gel filtration chromatography, dimer formation was found in a significant fraction of galectin-3 CRD (Wang et al., 1993). Therefore, whether this dimer interface is exclusively found for prototype galectins like galectin-l and galectin-Z-needs further examination. Galectin-3, a chimera-type galectin, contains a proline-glycine-rich domain. Galectin-3 contains a proline-glycine-rich domain at the NHz-terminal end (J ia and Wang, 1988), in addition to a CRD. This N-domain is composed mainly of sequence repeats, each consisting Of nine amino acids (PGAYPGXXX) in mammalian homologues (J ia and Wang, 1988) and eight amino acids (PGPYPGGP) in the chicken counterpart (Nurminskaya and Linsenmayer, 1996). So far, the role(s) of this domain is not clear, although similar features have also been seen in some other proteins, like a spliceosome- associated protein (SAP) SAP62 (Bennet and Reed, 1993) and an autoantigen annexin XI (Misaki et al., 1994). SAP62 is a defined component of spliceosome complexes E, A and B (Bennett et al., 1992; Hong et al., 1997). This splicing factor contains a C-domain of 22 proline- glycine-rich heptapeptide repeats with the consensus sequence PGVHPPA (Bennett and Reed, 1993) besides the Zn fmger-containing N-domain. It has been proposed that the P-, G-rich domain may be involved in protein-protein interaction (Bennett and Reed, 1993). Nevertheless, it will be interesting to see how it is related to galectin-3 N-domain as both SAP62 and galectin-3 are involved in pre-mRNA splicing. Annexin XI, on the other hand, was identified as a calcyclin-associated protein (Tokumitsu et al., 1992), which is recognized by sera of patients with various autoimmune diseases (Misaki et al. , 1994). Structurally, it contains a proline-glycine-rich 9 N-terrninal tail domain as well as a core domain that was conserved among annexin family. The N-terrninal tail has been shown to be responsible for its nuclear localization (Mizutani et al., 1995). Interestingly, annexin XI, a 56-KDa protein also shows a dual (nuclear and cytoplasmic) localization (Misaki et al., 1994), which raises the possibility that galectin-3 N-domain may also have the same intracellular targeting capacity. INTRACELLULAR SORTING Subcellular localization of galectins has been extensively studied for galectin-1 and galectin-3. These two galectins, like a large number of other proteins (Smalheiser, 1996), have been found to reside in multiple cellular compartments. Protein targeting between different compartments is usually via signal-mediated pathways. It is always interesting to see how different signals are incorporated into the protein structure, and how a particular signal is selected over others. Localization studies on galectins definitely may help answer these questions, considering their relatively simple primary structures. In this section, I will concentrate on the pathways mediating the extracelluar secretion as well as nucleocytoplasmic distribution. Galectin-l and galectin-3 are secreted out of cells via the pathway(s) distinct from the classical pathway. Most extracellular proteins leave the cells using the classical secretory pathway, which requires a signal peptide on the substrate proteins (Rothman and Orci, 1992). The secreted proteins enter the ER, and move to the cell surface via the Golgi apparatus. Both galectin-1 and galectin-3 lack a classical secretory signal (Muesch et al., 1990) and are secreted through so-called non-classical secretory 10 pathway(s). Examples of proteins utilizing nonclassical pathways include interleukin-113 (Siders and Mizel, 1995), FGF-l (Tarantini et al., 1995), and FGF-2 (Piotrowicz et al., 1997). Both galectin-l and galectin-3 have been used as a model system to explore these pathways. Galectin-l appears to be externalized from cells through extracellular vesicles derived from evaginations of plasma membrane. Galectin-l has been found to be secreted at a higher rate in differentiated muscles cells than in myoblasts (Cooper and Barondes, 1990). Immunofluorescent localization studies showed galectin-l is primarily cytoplasmic in myoblasts, but cytoplasmic staining is largely lost in myotubes, the well-differentiated muscle cells. Examination of differentiating myoblasts, the intermediate between two stages, showed that galectin-1 had a patchy distribution at this transition. These patches were in the outermost layer of cytoplasm and appeared to correspond to the evaginations in plasma membranes. Cooper and Barondes (1990) hypothesized that these evaginations may later mature into extracellular vesicles and serve as the secretory vehicles for galectin-1. Yeast cells can also secret galectin-l , when they are transformed with a galectin- l-expressing vector (Cleves et al., 1996). This Observation indicates that a pathway for secreting galectin-1 is conserved between budding yeast and mammalian cells. This secretion does not require any components in the classical secretory pathway nor the yeast multidrug resistance-like protein Ste6p, an a factor transporter. A genetic screen was designed to identify gene products which are involved in secretion. Two gene products, NCEl and NCE2, are able to enhance galectin-l secretion when they are ll overexpressed. (Cleves et al., 1996). Furthermore, nce2 null mutants showed a great reduction in galectin-1 secretion, establishing the pivotal role of NCE2 in nonclassical pathway for galectin-1. The primary structure of NCE2 may allow the polypeptide to fold into a membrane protein. Whether this protein is related to the secretion mechanism seen in muscle cells awaits more documentation. Galectin-3 is selectively secreted from apical surface, but not from the basolateral membrane, of Madin-Darby canine kidney (MDCK) cells via a nonclassical pathway. Plasma membranes of polarized epithelia cells have two domains, the apical and basolateral, which are separated from each other by tight junctions (Sirnons and Wandinger-Ness, 1990; Mostov et al., 1992). Distinct sets of proteins are targeted to each domain. Galectin-3 in MDCK cells is specifically secreted from the apical surface through a nonclassical pathway (Lindstedt et al., 1993). This conclusion is supported by the observation that this pathway was not inhibited by brefeldin A or monensin, drugs that inhibit the ER-Golgi pathway. On the other hand, the secretion is enhanced by calcium ionophore A23187 and by heat shock at 42°C. Lowered temperature as well as addition of nocodazole blocks its secretion. All of these distinguish this pathway from the classical pathway. Galectin-3 distributes between cytoplasm and the nucleus as a function of proliferating capacity. In mouse 3T3 fibroblasts, galectin-3 was found both in cytoplasm and in the nucleus. (Moutsatsos et al., 1986). The distribution between these two compartments correlates with proliferative states of cells (Moutsatsos et al., 1987). While in serum-starved cells, galectin-3 is primarily cytoplasmic, it becomes 12 predominantly nuclear as cells are reactivated by serum addition. Similar results can be seen in dense cultures versus sparse cultures. Sulicellular fractionation studies showed that galectin-3 in nuclear fraction has two isoforrns, a pI 8.2 phosphorylated form and a pl 8.7 dephosphorylated form. However, only phosphorylated isofonn is seen in cytosolic fraction. These results suggest that phosphorylation/dephosphorylation is involved in distribution between two compartments. Similar results have also been shown on human primary fibroblasts. These cells are susceptible to senescent change (Hamann et al., 1991; Cowles et al., 1989). In young cells, galectin-3 is mainly in the nucleus. As cells become senescent, galectin-3 is exclusively found in cytoplasm. Altered intracellular distribution has also been seen in neoplastic cells (Lotz et al., 1993). Paradoxically, galectin-3 is exclusively cytoplasmic in colon carcinoma cells, in contrast to its predominant nuclear localization in well-differentiated epithelial cells. Nuclear galectin-3 is associated with the nuclear matrix defined by detergent and high-salt extraction. In immunofluorescence studies, galectin-3 was found diffusely distributed throughout the nucleoplasm excluding nucleoli. Even after detergent and high-salt extraction, it still associates with nuclear structures that contain splicing factors SC35 and Sm proteins (Wang et al., 1995a; Hubert et al., 1995; Vyakarnam et al., 1997). The association is dependent on RNA-containing structures. Consistent with these results, galectin-3 has been found in purified nuclear matrix of adenocarcinoma cells (Wang et al., 1995b). Ultrastructurally, galectin-3 is found in nucleoplasm, interchromatin spaces excluding interchromatin granules, and on the border of condensed chromatins (Hubert et l3 al.,l995). The latter is considered as the site of mRNA synthesis and early events of pre- mRNA splicing (Spector, 1996). Therefore, this result agrees with the notion that galectin-3 is a pre-mRNA splicing factor. Most surprisingly, galectin-3 is also associated with dense fibrillar components and periphery of nucleoli, in contrast to the exclusion of galectin-3 fi'om the nucleoli under fluorescence microscope (Hubert et al., 1995). What causes this discrepancy is still not clear. The import of galectin-3 into the nucleus has been studied and preliminary data showed that galectin-3 probably enters the nucleus through an active mechanism. Transfection experiments revealed that FLAG-tagged constructs expressing the duplicate repeat and triplicate repeat of the galectin-3 coding sequence can enter the nucleus as well as FLAG-tagged galectin-3 (Tsay and Wang, unpublished results). Inasmuch as both duplicate and triplicate repeats have molecular sizes exceeding the diffusion limit of nuclear pores, about 40~60 kDa for globular proteins (Peters, 1986), we concluded that some active mechanisms are required for the entry for these constructs. Whether there exists an import signal for galectin-3 itself still awaits further investigation. Galectin-l is also present in nuclear structures that contain splicing factors. Confocal microscopic experiments showed that galectin-l is also localized in nuclear structures with Sm proteins as well as SC35, which supports the notion that it participates in pre-mRNA splicing (V yakarnam et al., 1997). It will be interesting to know whether its association with these structure is also dependent on RNAs. If the association is also sensitive to RNase treatment, this implicates that the CRD is probably responsible for the binding activity for both galectin-1 and galectin-3. However, if no RNA dependence is l4 seen for galectin-1, it will support the notion that the N-domain of galectin-3 perhaps plays an important role in association with nuclear matrix. BIOLOGICAL FUNCTIONS Galectins were initially found in the quest for proteins that recognize cell surface carbohydrates, based on the presumption that they perhaps participated in cell-cell interactions (Barondes, 1997). However, there is accumulating evidence indicating that they have many additional functions, both intracellularly and extracellularly. Whereas most activities are related to their saccharide binding activity, some are not necessarily dependent on this characteristic property. Galectins are involved in programmed cell death. Galectin-l has been found to induce apoptosis in activated T cell, (Perillo et al., 1995) and thymocytes (Perillo et al., 1997). Apoptosis is an important mechanism for the production of irnmunocompetent cells and for the termination of an immune response. In activated T cells, galectin-1 induces apoptosis via a Pas-independent pathway, and CD45 appears to be the cell surface receptor that interacts with galectin-1. N-glycans on CD45 may be essential for galectin-l binding, since the induction by galectin-l can be inhibited by swainsonin, an inhibitor for N-linked oligosaccharide processing (Perillo et al., 1995). Because most of galectin-1 is in dimeric form at the concentration (10 11M) that induces apoptosis, dimeric formation was proposed as a requirement to elicit the reaction (Perillo et al., 1995). Moreover, susceptibility to galectin-l-induced apoptosis is cell cycle- 15 dependent. Thymocytes at G1 phase are less sensitive to apoptotic change induced by galectin-1 than at other phases (Perillo et al., 1997). A tandem-repeat type galectin, galectin-9, also was found to induce cell death in thymocytes (Wada et al., 1997). The induction is cell type-specific, since hepatocytes are resistant to the stimulation. Lactose can suppress the response, suggesting that sugar- lectin interaction is involved in this process. Galectin-3 did not elicite simliar response on thymocytes (Wada et al., 1997). In contrast, it protects leukemic T cells from the apoptotic response induced by anti-Fas antibody and staurosporine (Yang et al., 1996). Interestingly, galectin-3 contains NWGR motif seen in the Bel-2 family, which is involved in heterodimer formation between Bel-2 (a suppresser of cell death) and Bax (a cytotoxic protein) (Yin et a1, 1994; Hanada et al., 1995). Irnmunoprecipitation analyses showed that galectin-3 indeed complexes with Bcl- 2 (Yang et al., 1996), but its significance in the apoptotic pathway remains to be determined Galectin-l participate in axon growth and guidance for olfactory neurons. Galectin-l is expressed by primary Olfactory neurons, olfactory nerve glial cell, and second-order neurons in the olfactory bulb (Puche and Key, 1995). In vitro, recombinant galectin-1 and its ligand can specifically stimulate neurite growth by cultured olfactory neurons, which were determined by increased neurite numbers as well as increased neurite lengths. (Puche et al., 1996). When intemeuron connection was examined in - transgenic null mice lacking galectin-1 (Poirier and Robertson, 1993), connections between neurons in the nasal cavity and the olfactory bulb were defective. A subpopulation of axons failed to navigate to their target site in the olfactory bulb (Puche 16 et al., 1996). This is the first phenotype ascribed to the lack of galectin-1 and indicates that galectin-1 has a role in the guidance of axon growth of olfactory neurons. Galectin-l and galectin-3 are pre-mRNA splicing factors. The observation that saccharide ligands for galectins inhibited splicing in a cell-free assay first implicated that galectins may participate in pre-mRNA splicing (Wang et al., 1991; Dagher et al., 1995). When galectins were selectively depleted from nuclear extracts of HeLa cells using lactose-agarose affinity adsorption, the splicing activity was lost, showing that galectins are essential for pre-mRNA splicing. For these galectin-flee extracts, either recombinant galectin-3 or galectin-l was able to restore its splicing capacity, and this restored activity is still subjected to saccharide inhibition (Dagher et al., 1995; Vyakamam et al. , 1997). This result indicated that either galectin-3 or galectin-1 is sufficient to complement the splicing activity and also showing the importance of CRD. The latter notion is further supported by the observation that C-domain of galectin-3 alone also reconstitutes the splicing activity (V yakarnam et al., 1997). When these two galectins were specifically removed from nuclear extracts by dual immunodepletion, the results were the same as those using lactose affmity adsorption. This argues that galectin- ] and galectin-3 are the two major galectins present in the nuclear extracts. How do galectins participate in the splicing pathway? Assembly of splicing substrates into splicing complexes in cell-free assay (figure 3) can be monitored by a ribonucleoprotein (RNP) gel electrophoresis system (Zilhnann et al., 1988). When splicing substrates were treated with galectin-free nuclear extract, only H complexes were Observed on the gel. When recombinant galectin-3 was added to restore the splicing activity, it also enhanced the formation of advanced spliceosomes including A complex Figure 3. Schematic representation of the involvement of small nuclear ribonucleoprotein particles (snRNP) and non-snRNP proteins in pre-mRNA splicing (modified from Moore et al., 1993). Pre-mRNA (top line), containing two exons separated by an intron, enters splicing complexes with snRNPs and exits as ligated mRNA (bottom line). Nascent pre-mRNA is bonnd by a multitude of hnRNP proteins immediately after transcription in vivo (Dreyfuss et al., 1993; Weighardt et al., 1996), forming H complex. E complex, the commitment complex in mammalian extracts, contains both U1 and U2 snRNPs, although U2 snRNP has a weaker affinity to the substrate at this stage (Michaud and Reed, 1991). Transition from E complex to A complex is rapid and requires ATP hydrolysis (Liao et al., 1992). The SR protein family of splicing factors are involved in this conversion (V alcarcel and Green, 1996). Then, U4/U6.U5 tri-snRNP complex enter spliceosome to form complex Bl. After structural arrangement, complex B1 is transformed into complex B2. Complex C1 carries out the cleavage at the 5' splice site and formation of the lariat intermediate, whereas complex C2 in the spliceosome immediately following exon ligation. 18 ——A3’-n PRE-mRNA + HnRNPs m—Al— HcomeLex U51 * *r 1121 ”.2" 1112“; m” —Ar Pyt— E COMPLEX SFSI:SF3b SC35 ATP L33 ca A COMPLEX B1 COMPLEX 32 COMPLEX C1 COMPLEX C2 COMPLEX 19 as well as B1/B2 and C1/C2 complexes (Dagher et al., 1995; Zillmann et al., 1988). This result implied that galectin-3 is at least required for the progression fiom H complex to A complex. It is not known whether depletion of galectins blocks the conversion from H to B complex, or from H to A complex (Figure 4). Considering that CRD alone is sufficient to restore the splicing activity of galectin-depleted nuclear extracts, it appears that carbohydrate-recognition domain already contains sufficient elements mediating the splicing reaction. A legitimate , question is whether carbohydrate-binding activity is required for splicing activity of galectins. Recent data show that the galectin-1 mutant without sugar-binding activity is still able to restore the splicing activity, although the saccharide inhibition is no longer observed (Dagher and Patterson, unpublished results). This indicates that sugar binding activity is, in fact, dispensable for splicing activity, arguing against the model that a sugar-lectin interaction is required for progression of spliceosome. A plausible explanation is that inhibition of splicing by saccharide perhaps reflects that the impact on the CRD conformation by saccharide binding. It has been shown that galectin-3 CRD have conformations of different thermal stabilities in the presence or absence of ligands (Agrwal et al., 1993). It is possible that these two conformations represent active and inactive forms for splicing activities of CRDs and lactose ligands act as a switch for their interchange. Without ligand, the active form participates in splicing reaction; with bound ligand, the active form becomes converted into the inactive form and thus CRD loses its splicing activity (Figure 4). Whether there exits a switch in intact cells will be another interesting issue. 20 Figure 4. Roles of galectin CRD in pre-mRN A splicing. Galectin CRD participates in the conversion from H complex to A complex (Dagher et al., 1995). Depletion of galectins by lactose-affmity adsorption or immunopresipitation inhibits the progression of spliceosome formation. On the other hand, saccharide ligand induces a conformational change on CRD structures, which results in inactivation of galectins and thus inhibition of splicing reaction. 21 m—A-PY—m l HnRNPs H COMPLEX IMMUNODEPLETION LACTOSE ADSORPTION ® ACTIVE + LACTOSE - LACTOSE --. flflflflflflfl SFSaISF3b A COMPLEX . INACTIVE 22 INTRODUCTION TO NUCLEAR TRANSPORT Nucleocytoplasmic transport plays a fundamental role in coordinating the functions of the nucleus and cytoplasm (Nigg, 1997; GOrlich and Mattaj, 1996). It is mediated by a large molecular structure that spans the nuclear envelope called the nuclear pore complex (Davis, 1995). Proteins that function in the nucleus are synthesized on free ribosomes in the cytoplasm and imported into the nucleus. RNAS that function in cytoplasm are transcribed and processed in the nucleus and exported into the cytoplasm (GOrlich and Mattaj, 1996). Both processes occur through nuclear pore complexes. In the following section, I will discuss the structure of nuclear pore complex, the pathways for nuclear import, as well as the general properties of nuclear export. NUCLEAR PORE COMPLEX Nuclear pore complexes (NPC) have a mass of 125 megadaltons and contain about 100 different polypeptides. Characteristic features of many vertebrate nucleoporins (nuclear pore proteins) are modifications with O-linked N- acetylglucosamine (GlcNAc) and the presence of short degenerate repeats, such as FXFG repeats and GLFG repeats. The NPC contains a passive diffusion channel about 9 nm in diameter (Peters, 1986). Small proteins with sizes less than 40-60 KDa can diffuse freely through the pore. Proteins above the diffusion limit can enter the nucleus only via an active mechanism. Nevertheless, small nuclear proteins generally enter the nucleus via active mechanism rather than by diffusion. 23 Central Plug Cytoplasmic Filament Cytoplasmic Ring ' . Lumenal ~ - Domain Nuclear Basket Terminal Ring Figure 5. Schematic respresentation of a consensus model of the membrane-bound nuclear pore complex. Nuclear pore complex contains four major elements: spoke-ring assembly, central plug, cytoplasmic filaments, and nuclear basket (Bastos et al., 1995). 24 F our basic elements of structure have been observed for nuclear pore complex (NPC) (Akey and Radermacher, 1993) (Figure 5). The main body is a spoke-ring assembly anchored on a specialized region of nuclear envelope. This structure consists of nuclear and cytoplasmic rings connected to each other by two sets of eight spokes . A central plug sits within the aqueous channel formed by the spoke-ring assembly. Peripheral components includes eight short cytoplasmic filaments that extend into the cytoplasm and nuclear basket that project into the nucleus. Many vertebrate nuclear pore proteins are O-linked glycoproteins that contains characteristic peptide repeat motifs. “A family of 10-20 nucleoporins has been identified as proteins that bind to wheat germ agglutinin (WGA) (Davis and Blobel, 1987). WGA binding is due to the extensive modification with O-linked N- acetylglucosarnine moieties on the NPC structure (Hart et al., 1988). cDNAs encoding six members (p62, POM121, NUP153, NUP214/CAN, NUP98 and NUP358/RanBP2) of O-linked glycoprotein family of nucleoporins have been isolated in vertebrates (Table 2). The deduced amino acid sequences showed that each member contains multiple copies of XFXFG repeats, where X is any amino acid with a small or polar side chain. This motif is also seen in a few yeast nucleoporins, like Nuplp (Bogerd et al. , 1994; Belanger et al. , 1994) and Nup2p (Belanger et al., 1994). A GLFG motif, which is fi'equently seen on yeast nucleoporins, has only been found on NUP98 in vertebrates. Immunoelectron microscopy has been used to determine locations of these glycoproteins within NPC. Namely, p62 is found both at cytoplasmic and nuclear sides (Panté et al., 1994); POM21 is on membrane domain of NPC (Hallberg et al., 1993); NUP358 as well as NUP214 are mapped to cytoplasmic filaments (Boer et al., 1997; Wu 25 Table 2. Vertebrate nuclear pore proteins Name Location O-linked glycoprotein family p62 POM121 NUP153 NUP2 14/ CAN NUP98 NUP358/ RanBP2 Others gp2 10 NUP107 NUP155 TPR/ p270 NUP180 NUP84 (NUP88) hCRMl (1 12kDa) Central region; both cytoplasmic and nuclear (Panté et al., 1994) Pore membrane domain Terminal ring of the nuclear basket (Panté et al., 1994) Cytoplasmic filaments (Panté et al., 1994) and nucleoplasmic face (Boer et al., 1997) Nucleoplasmic side Cytoplasmic fibers Pore membrane domain ? Both nuclear and cytoplasmic Cytoplasmic (7) and nuclear filaments (Cordes et al., 1997) Cytoplasmic ring and filaments Cytoplasmic face Properties and possible functions XF XF G repeats at N-tenninus and heptad repeats/coiled-coil structure at C-domain.; complexed with p58 and p54; essential for NPC function; multiple O-GlcNAc and sialic acids (Snow et al., 1987; Starr et al., 1990; Emig et al., 1995; Buss et al., 1995)) XFXF G repeats at C-terminus; integral membrane protein; membrane anchor; multiple O-GlcNAc (Hallberg et al, 1993) XFXFG repeats at C-tenninus; four Zn fingers; homodimerization; multiple O-GlcNAc ; involved in mRNA export (Sukegawa and Blobel, 1993; Bastos et al., 1996)) XFXF G repeats at C-terminus; leucine zipper motif; complexed with p75; putative oncogene; docking site for karyopherins (Kraemer et al., 1994) GLFG, FG, FXFG repeats at N-terminus; docking sites for karyopherins (N-terminus); related to DNA replication; multiple O-GlcNAc; involved in RNA export but not protein import (Powers et al., 1995; Radu et al., 1995; Powers et al., 1997) XFXFG, F G motifs; four Ran binding sites; leucine- rich domain; cyclophilin A homologous domain; docking sites for karyopherins and Ran (Y okoyarna et al., 1995; Wu et al., 1995) Type I integral membrane protein; membrane anchor; high mannose N-linked oligosaccharides on lumenal domain (Gerace et al., 1982; Wozniak et al., 1989; Greber et al., 1990; Wozniak and Blobel, 1992) Leucine zipper at C-terminus (Radu et al., 1994) Multiple potential phosphorylation sites (Radu et al., 1993) Coiled-coil motif at central domain; activation of oncogenic kinases (Byrd et al., 1994) Recognized by serum form patients with autoimmune diseases (Wilken er al., 1993) Coiled-coil domain at C-terminus; bound with Nup214; non-glycosylated; no repeat motif; CAN- dependent NPC localization (Bastos et al., 1997; Fomerod et al., 1 177)) Localized at pores and nucleoplasm; karyopherin 13- like domain (Fomerod et al., 1997) 26 et al., 1995); and NUP98 and NUP153 are on the nuclear side of NPC (Panté et al., 1994; Radu et al., 1995). In other words, nucleoporins with XFXF G repeats can be along the cytoplasmic-nuclear axis of NPC. The O-linked glycoproteins are essential docking sites for nuclear transport. The majority of these proteins bind to the karyopherins, a family of transport factors, directly, and have be proposed to form an array of sites for mediated docking of transport substrates (figure 6) (Radu et al., 1995; Wu et al., 1995). For example, NUP358 has been considered as the site where protein import complex is initially assembled (Wu et al., 1995), containing docking sites for Ran GTPase as well as karyopherin Oil/01243 complexes. In fact, more and more data suggest that anchoring sites on these nucleoporins are shared by all of the nuclear import pathways and export pathways. This is exemplified by the fact that the B—type karyopherins are all capable of binding to these glycoproteins, and their binding is competitively inhibited by each other (Bonifaci et al. , 1997; Yaseen and Blobel, 1997). Moreover, it was recently shown that all of the signal- mediated pathways were inhibited, when these glycoproteins in NPC were masked by mutants of karyopherin Bl (Kutay et al., 1997). Other nucleoporins. Sequence information is also available for several other vertebrate nucleoporins (Table 2). None of them contains XFXFG or GLF G repeats nor have O-linked GlcNAc. Some contain the sequence motifs that are likely to mediate protein-protein or protein-nucleic acid interactions, such as zinc fingers, leucine zippers and coiled-coil motifs. 27 NUP358* NUP214* NUP84 NUP180 gp210 * NUP155 P62 Pomzl‘ NUP214* NUP98* Tpr/p27 * NUP153 Figure 6. Diagram summarizing localizations of characterized nucleoporins within the nuclear pore complex. Nucleoporins that have been shown to bind to karyopherin complexes are placed at the right side. These proteins may serve as docking sites as import complexes move along the nuclear pore. Asterisk indicates nucleoporins that have XFXFG repeats as well as O-linked GlcNAc modifications. 28 Most yeast nucleoporins also have characteristic repeat motifs seen in vertebrate homologues but are not modified by sugar residues. Some yeast nucleoporins contains XFXFG repeats that are the signature motif of NPC O-linked glycoproteins of vertebrates. These yeast proteins, however, appear not to be modified by GlcNAc (Kalinich and Douglas, 1989), which is consistent with the fact that WGA does not block nuclear protein import in in vitro import assay (Kalinich and Douglas, 1989). A second family of nucleoporins is characterized by the repeat GLFG. It appears that yeast and vertebrate NPCs have some differences, considering that the basic structure units and posttranslational modifications are not entirely identical. NUCLEAR IMPORT As mentioned, molecules with sizes above the exclusion limits (40-60 kDa) primarily enter the nucleus in an active way. Usually, this kind of transport is selective and signal-dependent. Proteins or RNAs bearing import signals are recognized by specialized receptors and then targeted to nuclear pores, where these molecules are translocated across the nuclear envelope. Accumulating evidence implies that specificity of a multitude of pathway arises at the earlier steps, i.e., signal-receptor recognition. Different import pathways appear to use the same set of nucleoporins to dock import substrates and translocate them across the nuclear pores (Bonifaci et al., 1997; Yaseen and Blobel, 1997). Therefore, I would like to discuss the import pathways based on the pairing between import signals and their receptors (Table 3). 29 Table 3. Signals and Receptors Involved in Nuclear Import Receptor Import Signal Example Sequence Substrates Karyopherin (11 Nuclear Localization Nuclear proteins (Importin-a/NPI-l Signal PKKKRKV SV40 T antigen SRPl/NTPCSS/ Classical signal KRPAAIKKAGQAKKKK Nucleoplasmin Kap60p) Bipattite signal Karyopherin (12 Nuclear Localization Nuclear proteins (Rchl) Signal PKKKRKV SV40 T antigen Classical signal KRPAAIKKAGQAKKKK Nucleoplasmin Bipattite signal Karyopherin a3 ? ? ? Karyopherin B IBB domain of RMRKFKNKGKDTAELRR Karyopherin or (Kap97p) karyopherin a RRVEVSVELRKAKKDE QILKRRNV Transportin M9 sequence NQSSNFGPMKGGNFGGR hnRNP A1 (Karyopherin B2 SSGPYGGGGQYFAKPR /Kap104p) NQGGY Karyopherin B3 ? ? Ribosomal proteins? ? KNS sequence YDRRGRPGDRYDGMVGF hnRNP K SADETWDSAIDTWSPSE WQMAY ? Pro, Gly-rich domain N-tenninus (aa 1-206) ANNEXIN XI Snurportin Trimethylguanosine cap U snRNAs 30 Karyopherin a/Bl (011 B1 and a2Bl) mediated pathway for common nuclear proteins is the best characterized nuclear import pathway. Most knowledge of this pathway originates from studies in Xenopus and mammalian systems. Experiments based on microinjection into Xenopus oocytes discovered the two steps of this import process: energy-independent docking to the NPC and energy-dependent translocation through the pore (Richardson et al., 1988; Newmeyer and Forbes, 1988). The major breakthrough was the development of an in vitro system based on cultured cells treated with digitonin to selectively permeabilize the plasma membrane (Adam et al., 1990). Digitonin- permeabilized cells retain the capacity to import nuclear proteins with the supplement of fractionated cytosol. From these cytosolic fractions, required factors for nuclear import were purified. Nuclear import of NLS-containing proteins has been distinguished as two steps: docking and translocation (Figure 7). First, the import substrates binds via its NLS to the karyopherin or, either 011 or 012, in the cytoplasm (GOrlich et al., 1994; Imamoto et al., 1995), which also augments the interaction between karyopherin or and B1 subunits (Moroianu et al., 1996). Karyopherin B1 contains binding sites with repeat- containing nucleoporins, and mediates the docking of the complex onto the nuclear pores in an energy-independent manner (Morioanu et al., 1995). The import complex is subsequently translocated through the pore by an energy-dependent mechanism (Richardson et al., 1988; Newmeyer and Forbes, 1988), which is probably achieved by sequential docking-undocking process mediated by Ran, a Ras-like GTPase (Melchior et al., 1993; Moore and Blobel, 1993; Kutay et al., 1997), and NTF2 (Moore and Blobel, Figure 7. Nuclear import of NLS-containing proteins has two distinct steps: docking and translocation. Docking requires only karyopherina and B, and does not need GTP. Translocation is mediated by Ran GTPase and its cofactor NTF2, which requires GTP hydrolysis. 32 1994; Paschal and Gerace, 1995). After being transported to the nuclear side of NPC, the import complex is dissociated. As the karyopherin B1 was promptly exported by an NBS-mediated pathway (Iovine and Wente, 1997), the karyopherin (Jr-substrate complex reaches the nucleoplasm (Moroianu et al., 1995). Ran GTPase cycle is coupled with the translocation of karyopherin complex. Like other GTPases, Ran has a set of effectors that modulate its GTPase activity. The only known guanine nucleotide exchange factor for Ran is a nuclear protein RCCl (Bischoff and Ponstingl, 1991); the only known Ran GTPase activating protein is cytoplasmic (Hopper et al., 1990; Bischoff et al., 1995). Based on distinct locations of these two effectors, it appears that GTP exchange occurs only in the nucleus and GTP hydrolysis is only in the cytoplasm. The Ran GTPase cycle. Koepp and Silver (1996) proposed that Ran moves out of the nucleus in a GTP-bound form and it enters the nucleus at the GDP-bound state (Koepp and Silver, 1996) (Figure 8A), assuming that Ran-GTP is enriched by nuclear RCCl in the nucleus and Ran-GDP is formed in cytoplasm due to augmented GTPase activity by RanGAP. This model also predicted that Ran-GDP moves toward cytoplasm and Ran-GTP heads for the nuclear side so that the cycle can be completed (Figure 8). Some of the experimental data are consistent this model: 1. Ran-GDP has been found to be required for nuclear import (Weis et al., 1996). 2. When RanGAP was microinjected into the nuclei, nuclear export of NES- containing protein was blocked (Richards et al., 1997). 33 Figure 8: (A) A model illustrating Ran GTPase cycle. Two effectors for Ran are at different compartments. RCCl is localized in the nucleus and RanGAP is in the cytoplasm. In order the complete the cycle, Ran-GDP flows toward the nucleus and Ran- GTP move toward cytoplasm. (B) Coordination of protein import by Ran GTPase cycle. Due to the compartrnentation of RCCl and RanGAP, Ran-GDP and Ran-GTP are enriched at different sides. Ran-GDP is more abundant at cytoplasmic side, where it promotes the docking complex for protein import. Ran-GTP is more abundant at the nuclear side, where it stimulates the dissociation of karyopherin-substrate complex. The net effect is that nuclear protein is transported from cytoplasmic side to the nuclear side. 35 3. GTP-bound Ran mutant lacking GTPase activity is still able to mediate nuclear export, indicating that Ran-GTP but not GTP hydrolysis directly participate in nuclear export (Richards et al., 1997). 4. When nuclear import was blocked by nonhydrolyzable GTP analogues, Ran was specifically localized at the cytoplasmic surface (Melchior et al., 1995). Current data indicates that Ran GTPase cycle coordinates the import pathway. It has been shown that karyopherin 01/ B1 and imported substrate form a stable complex in the presence of GDP-bound Ran, but karyopherin a-ligand is released from the complex by Ran-GTP (Rexach and Blobel, 1995; Kutay et al., 1997). Since the Ran-GTP is mainly nuclear and Ran-GDP is enriched in cytoplasm according to the model, the Ran- GDP mediates the import complex formation at the cytoplasmic side and Ran-GTP will dissociate the substrate from karyopherins in the nucleus (Figure 8B). In summary, the distinct distribution of RCCl and RanGAP across the nuclear envelope enriches GDP- and GTP-bound forms of Ran at cytoplasmic side and nuclear side respectively. This, in tum, potentially helps to establish the unidirectional movement of nuclear proteins (Figure 8B). NTF2 assists the docking of Ran-GDP as well as import complexes to the repeat-containing nucleoporins. NTF2 was first identified as a required factor for nuclear import, complementing the functions of Ran (Moore and Blobel, 1994). In yeast, NTF2 has specific affinity to nucleoporins containing FXFG repeats, like Nuplp, Nsplp, Nupl45p , Nup57p and Nup36p. Stronger binding with some of these nucleoporins, like Nup36p requires the presence of Ran-GDP, Kap95B and Kap60a. Therefore, NTF2 has 36 been proposed as the regulator that coordinates Ran-dependent association and dissociation reactions underlying nuclear import (Nehrbass and Blobel, 1996). A similar interaction between karyopherins, Ran, nucleoporin, and NTF2 has also been observed in vertebrate cells (Paschal and Gerace, 1995). Transportin-M9 sequence mediated pathway is responsible for the nuclear import of hnRNP Al and its related proteins. HnRNPs are a set of proteins that bind to hnRNAs. They are involved in many aspects of RNA metabolism, including RNA processing and transport (Dreyfuss et al., 1993). In hnRNP A1, there exists a stretch of amino acids, designated as M9 (Siomi and Dreyfuss, 1995), that is responsible for nuclear shuttling property of hnRNP A1 (Piflol-Roma and Dreyfuss, 1993). Recently, its nuclear import receptor has been identified in a variety of organisms. In vertebrates, it was designated as transportin (Pollard and Dreyfuss, 1996), MIP (M9 Interacting Protein) (Fridell et al., 1997), and karyopherin B2; in yeast, it is named as Kap104p (Aitchison et al., 1996). This receptor is homologous to karyopherin B1 and specifically mediates nuclear import of hnRNP Al, but not NLS-containing proteins (Pollard et al., 1996; Fridell et al., 1997). Transportin, like karyopherin B1, binds to a set of nucleoporins containing the characteristic peptide repeat motifs. It mediates the docking of hnRNP A1 onto the pore, but translocation also requires Ran GTPase (Bonifaci and Blobel, 1997). There are a multitude of nuclear import pathways yet to be defined. For example, KNS sequence in hnRNP K is responsible for its nuclear shuttling property, and, interestingly, this sequence can dock proteins to the nuclear pore without the supplement of cytosolic factors (Michael et al., 1997). What nucleoporin mediates its 37 docking still remains to be studied. Some other potential receptor proteins are also identified, like karyopherin 1313 (Takeda et al., 1997) and karyopherin B3 (Yaseen and Blobel, 1997), but no substrates have been characterized yet. There is also a report on the identification of the receptor that mediates nuclear import of U snRNAs, which has a trirnethylguanosine cap that interacts with a receptor, snurportin (Lilhrmann, 1997). This receptor mediates nuclear import of U snRNPs in oocyte cells but only plays a minor role in somatic cells (Marshallsay and Ltihrmann, 1994). NUCLEAR EXPORT The list of signals and receptors involved in nuclear export is relatively small, compared to that for nuclear import (Table 4). Discovery of the cap-binding complex (Izaurralde et al., 1995) provided the first chance to examine the molecular mechanisms underlying RNA export and to explore the interplays between nuclear import and export. Nuclear export signals have also been recently identified in several nuclear shuttling proteins (Gerace, 1995). Cap-binding complex (CBC) is involved in RNA export. Monomethylated cap structures have been found to participate in several steps of RNA metabolism, including nuclear export (Harnm and Mattaj, 1990), initiation of translation (Sachs et al., 1997) and RNA processing (Izaurralde et al., 1994). Microinjection of cap analog, the dinucleotide m7GpppG, was shown to specifically inhibit nuclear export of U1 snRNA. For mRNAs, a monomethylated cap conferred a higher export rate than a dimethylated or trimethylated cap structure (Harnm and Mattaj, 1990; Jarmolowski et al., 1994), although the 38 Table 4. Signals and Receptor Involved in Nuclear Export Receptor Export signal Example Sequence Substrate Cap-binding 7-methylguanosine cap mGpppG, mGpppA U snRNAs; mRNAs complex hRIP Nuclear Export Signal LPPLERLTL (Rev) Nuclear proteins (NES) LALKLAGLDI (PKI) ? M9 sequence NQSSNFGPMKGGNFGGR hnRNP A1 SSGPYGGGGQYFAKPR NQGGY ? KNS sequence YDRRGRPGDRYDGMVG hnRNP K FSADETWDSAIDTWSP SEWQMAY 39 monomethylated cap was not essential for their export (Izaunalde et al., 1997). A cap- binding complex (CBC) was identified from nuclear extracts, which consists of two subunits: CBP20 and CBP8O (Izaurralde et al., 1992; Izaurralde et al., 1995). Antibodies raised against recombinant CBP20 indeed inhibited the nuclear export of U snRNAs, demonstrating that this complex is an nuclear export factor for RNA (Izaurralde et al., 1995). Although CBC seems not to be required for export of mRNAs, it does accompany mRNPs through nuclear pores on a particular pre-mRNA particle, the Blabiani ring (BR) RNP. BR particle is the product of the Balbiani ring (BR) gene in the salivary glands of the dipterin Chironomus tentans (Visa et al., 1996), whose giant size (35-40 kb) makes it possible to be visualized under electron microscope. CBC binds to the BR transcript as it was transcribed, and accompanies this RNP particle to travel through the nucleoplasm until it meets nuclear pores. During translocation through the channel, CBC remains at the 5’ end of the particle and is not detached until the RNA molecule extends into the cytoplasm (Figure 9) (Visa et al., 1996). Thereafter, it appears to be released from the BR RNP immediately, probably reentering the nucleus. Recent data showed that karyopherin a-Bl pathway is responsible for the nuclear import of this NLS-containing CBC (GOrlich et al., 1996). Nuclear export signals (NES) in certain nuclear shuttling proteins are responsible for their rapid exit from the cell nucleus. Nuclear export signals have been identified in several nuclear shuttling proteins. They were initially found in the HIV-1 Rev protein (Wen et al., 1995; Fischer et al., 1995) and in the Protein kinase A 40 BRRNP ____.. —> 40—. ._____+ 6,, Figure 9: The nuclear export of Balbiani Ring RNP particle. Three pre-mRN A splicing factors, hrp45, CBC20 and hrp36, travel with the transcript to different locations (modified form Danholt (1 997)). 41 inhibitor (PKI) (Wen et al., 1995). Both are short sequences and enriched in hydrophobic residues, especially leucines, with particular spacing. NBS-mediated export is energy and temperature dependent and confers a rapid export property to heterologous proteins (Wen et al., 1995). A receptor that interacts with this group of signals has been identified. The NES receptor, hRIP (human Rev Interacting Protein) or RAB (Rev Activation domain Binding protein), is a 59-kDa nucleoporin-like protein with FG peptide repeat motifs (Fritz et al., 1995; Bogerd et al., 1995). It is localized in the nucleus as well as the nuclear envelope (Fritz et al., 1995), and probably also cytoplasm (Bogerd et al., 1995). Known nuclear proteins containing NES include ( 1) components for nuclear transport of macromolecules, like Rev (Wen et al., 1995), Rex (Bogerd et al., 1996; Kim et al., 1996), karyopherin B1 (Iovine and Wente, 1997), and RanBPl (Zolotukhin et al., 1997), and (2) proteins involved in signal transduction pathways, like PKI (Wen et al., 1995), and MAPK kinase (Fukuda et al., 1996). M9-mediated pathway is related to nuclear export of mRNAs. Using a heterokaryon cell system, it has been shown that hnRNP Al has a shuttling capacity across nuclear envelope (Pifiol-Roma and Dreyfuss, 1993). The same conclusion is derived from the observation on its yeast homologue Npl3p (Flach et al., 1994). A domain was found to mediate the nuclear import and export activities, called M9 sequence (Siomi and Dreyfirss 1995; Michael et al., 1995). Although transportin is considered as M9 receptor, it seems not to be responsible for its nuclear export (Pollard et al., 1996; Bonifaci et al., 1997). When hnRNP Al was microinjected into either cytoplasm or the nucleus, it indeed saturates the export pathway for mRN As, but not for 42 U snRNAs (Izaurralde et al., 1997) This inhibition is independent of M9 sequence and may be exerted in cytoplasm, and the underlying mechanism is not clear at present. C tentans hrp36, a homologue of hnRNP Al, is a component of BR RNP particle (Visa et al., 1996). Its localizaiton has been studied using immunoelectron microscopy. This study showed that it was added to BR RNA concomitant with transcription, and remained on this RNP particle, while traveling through nucleoplasm. After leaving the nucleus, hrp36 was still present in this RNP even when polysomes were assembled on BR particles (Visa et al., 1996). Although hnRNP A] has extensive association with RNAS, direct evidence indicating that it is essential for mRN A export is still lacking. A novel signal conferring nuclear shuttling activities is found in hnRNP K. This KNS sequence, like M9 sequence, is responsible for the nuclear export as well as nuclear import property of hnRNP K1 , as demonstrated in a heterokaryon system (Michael et al., 1997). The molecular mechanism for its export awaits further investigation. Some of the pre-mRNA splicing factors are exported with processed transcripts. Both hnRNP A1 (Mayeda et al., 1992; Caceres et al., 1994) and CBC (Izaurralde et al., 1994; Lewis et al., 1996) are well documented pre-mRNA splicing factors. Although both of these proteins travel out of the nucleus with mRNAs, the timing of their release form the RNP is quite different. CBC appears to be released from the RNP complexes immediately following their arrival at cytoplasmic side of nuclear pores (Visa et al., 1996a), while hnRNP A1 probably accompanies RNAs from transcription to translation (Visa et al., 1996b). However, not every pre-mRNA splicing factor is exported with mRNP complex. For example, the hrp45 protein, a SR protein of 43 C. tentans, is found to associate with nascent RNAs concomitant with their synthesis via the binding with the exon. It is released from BR RNP particles in the nucleoplasm when the particle make contacts with the nuclear pores (Alzhanova-Ericsson et al., 1996) (Figure 9). Briefly speaking, different splicing factors seem to have distinct fates for binding with the pre-mRNA/mRNA particles. Based on distinct timing of release, this process seems to be a regulated process. How and why their binding with mRNPs is terminated will be interesting subjects to explore. CHAPTER II NUCLEAR EXPORT OF GALECTIN-3 IN MOUSE 3T3 FIBROBLASTS: I. PARAMETERS OF THE TRANSPORT IN DIGITONIN-PERMEABILIZED CELLS Yeou-Guang Tsay, Nancy Y. Lin, and John L.Wang Department of Biochemistry, Michigan State University East Lansing, MI 48824 44 45 SUMMARY Galectin-3 is a galactose-Ilactose-binding protein (M, ~30,000), identified as a required factor in the splicing of pre-mRN A. Immunofluorescence staining revealed that galectin-3 distributes differentially between the nucleus and the cytoplasm, depending on the proliferative state of the cells under analysis. Using digitonin-permeabilized mouse 3T3 fibroblasts, we provide evidence that galectin-3 is rapidly and selectively exported from the nucleus. Although both phosphorylated and nonphosphorylated isoforms of galectin-3 are found in the nuclear fraction, only phosphorylated galectin-3 is identified in the exported fraction, implying that phosphorylation is important for the nuclear export of the protein. In addition to galectin-3, we found that several other proteins are also rapidly exported in the digitonin-permeabilized cell assay. These include galectin-l, Ran, and karyopherin-B. In contrast, the Sm B and U1 A polypeptides of the small nuclear ribonucleoprotein complexes (snRNPs) are retained in the nuclear residue during the course of the same assay. The behavior of heterogeneous nuclear RNP (hnRNP) protein A1 is somewhat intermediate, exhibiting very slow export. The rate of galectin-3 export is decreased by cold temperature and by the addition of wheat germ agglutinin. More strikingly, galectin-3 export can be inhibited, at least partially, by the simultaneous addition of a peptide bearing a nuclear export signal plus a dinucleotide cap analogue found at the 5’-end of mRNAs. These results suggest that galectin-3 may be exported in association with RNP(s) containing monomethylated cap structure as well as polypeptide(s) bearing nuclear export signals. 46 INTRODUCTION Galectin-3 belongs to a family of widely distributed proteins that: (a) bind to B- galactosides; and (b) contain characteristic amino acid sequences in the carbohydrate recognition domain of the polypeptide (for reviews, see Barondes et al., 1994; Kasai and Hirabayashi, 1996; Leffler, 1997). Of the presently known 10 members of this family, galectins-1 and -3 have been shown to be factors involved in the splicing of pre-mRNA, assayed in a cell-free system (Dagher et al. 1995; Vyakamam et al., 1997). This conclusion is based on several key findings: (i) nuclear extracts (NE) derived from HeLa cells, capable of carrying out splicing, contain galectins-1 and -3; (ii) saccharides that bind the galectins with high affinity inhibit the cell-free splicing reaction; (iii) depletion of both galectins from NE, either by lactose affinity adsorption or by double antibody adsorption, resulted in the concomitant loss of splicing activity; (iv) depletion of either galectin-1 or galectin-3, by specific antibody adsorption, failed to remove all of the splicing activity and the residual activity was still sensitive to saccharide inhibition; and (v) either galectin-1 or galectin-3 alone is sufficient to reconstitute, at least partially, the splicing activity of NE depleted of both galectins. All of the results suggest that the activities of galectin-1 and galectin-3 in the nucleus may be redundant (Patterson et al. , 19971 A number of lines of evidence have been accumulated to indicate that galectin-3 is found in both the cytoplasm and nucleus of cells. Immunofluorescence staining, using 47 monoclonal, as well as polyclonal, antibodies specifically directed against galectin-3, was carried out on formaldehyde fixed, Triton X-100 permeabilized 3T3 fibroblasts. There was prominent labeling of the nucleus and variable staining of the cytoplasm (Moutsatsos et al. 1986; Wang et al., 1995). This dual localization of galectin-3, in the nucleus and cytoplasm, has been confirmed by immunoelectron microscopy (Hubert et al., 1995). Moreover, the nuclear staining of galectin-3, at the light and electron microscopic levels, was sensitive to treatment of the cells with ribonuclease A, but not to parallel treatment with deoxyribonuclease I, implying galectin-3 is associated with RNA-containing nuclear structures. Finally, the predominance of nuclear versus cytoplasmic distribution of galectin-3 depended on the proliferative state of the cells: sparse, proliferating cultures showed nuclear localization of galectin-3 whereas confluent, quiescent cultures yielded mostly cytoplasmic staining (Moutsatsos et al., 1987). Galectin-3 expression and its intracellular distribution have also been shown to vary along the crypt-tO-surface axis of human colonic epithelia (Lotz et al., 1993). The protein is concentrated in the nuclei of differentiated colonic epithelial cells. The progression from normal mucosa to adenoma to carcinoma is characterized by a paradoxically distinct absence of galectin-3 in the nuclei of adenoma and carcinoma cells. These observations on the dual localization of galectin-3 raise the possibility that the protein might shuttle between the cytoplasm and nuclear compartments. The rapidly expanding list of proteins that exhibit this nucleocytoplasmic shuttling property includes: (i) nucleolar proteins nucleolin and B23/No38 (Borer et al. , 1989); (ii) heat shock protein hsp70 (Mandell and Feldherr, 1990); (iii) transcription factors such as the steroid 48 receptors (Guiochon-Mantel et al., 1991; Chandran and DeFranco, 1992) and TFIIIA (F ridell et al., 1996); (iv) proteins of the heterogenenous nuclear ribonucleoprotein complex (hnRNP) such as hnRNP A1, A2, E (Pifiol-Roma, and Dreyfuss, 1993) and hnRNP K (Michael et al., 1997); (v) proteins involved in nucleocytoplasmic transport, including karyopherin-/importin-B (Iovine and Wente, 1997), Ran-binding Protein ' (Richards et al., 1996), the HIV Rev protein (Wen et al., 1995; Fischer et al., 1995), and Np13p (F lach et al., 1994; Lee et al., 1996); and (vi) various signal transduction proteins and their regulators, such as the heat stable inhibitor of CAMP-dependent protein kinase (PKI) (Wen et al., 1995) and mitogen-activated protein kinase kinase (F ukuda et al., 1996) Studies on nuclear protein import have identified some of the key components that function in recognition, docking, and translocation steps of the process. Karyopherin-/importin-Or is one subunit of the receptor that recognizes the nuclear localization signal (NLS) and, along with karyopherin-B, mediates the docking of import- competent substrates to the nuclear pore complex (GOrlich et al., 1994; Radu et al., 1995). A GTP-binding protein Ran/TC4 and the associated factor, plO/NTF 2, are required for the translocation of the substrate proteins through the nuclear pore complex (Moore and Blobel, 1993; Melchior et al., 1993). These advances in our understanding of nuclear protein import have been possible mainly as a result of the development of an in vitro assay, using digitonin-penneabilized cells (Adam et al., 1990). This assay takes advantage of the fact that digitonin treatment of cultured cells penneabilizes the plasma 49 membrane to macromolecular substrates while retaining the structural integrity of the nuclear envelope. Thus, the nuclei retain the ability to transport and accumulate proteins bearing the appropriate NLS. The system is particularly amenable to fractionation and biochemical analysis of the components involved in the transport process. We have taken advantage of this digitonin-permeabilized cell system to test the possibility that galectin-3 might be exported from the cell nucleus, a requirement for a nuclear protein to shuttle between the nucleus and the cytoplasm. In the present communication, we report that, indeed, galectin-3 is rapidly and selectively exported from the nucleus of digitonin-permeabilized cells. In the accompanying paper, we report the results of our initial characterization of the exported components. 50 EXPERIMENTAL PROCEDURES Cell Culture and Reagents NIH mouse 3T3 fibroblasts were Obtained from the American Type Culture Collection (Rockville, MD). The cells were grown as monolayers in Dulbecco’s Modified Eagle Medium containing 10% calf serum, 100 U/ml penicillin, and 100 ug/ml streptomycin at 37 °C in a humidified atomsphere of 10% C02. Routinely, the cells were used in the export assay at a density of ~ 1.5 x 10‘ cells/cmz, about 30% of the saturation density. The nuclear export assay was also carried out using 3T3 cells metabolically labeled with 32PO4 (New England Nuclear, Boston, MA; 50 uCi/ml culture medium, 5 ml/ culture dish). A rat monoclonal antibody was developed against the Mac-2 antigen (Ho and Springer, 1982), which has been shown to be galectin-3 (Cherayil et al., 1989). The antibody was prepared from serum-free cell culture supernatant derived from the hybridoma line as described (Wang et al., 1995); this antibody preparation is hereafter designated as rat anti-galectin-3. The polyclonal rabbit antiserum against recombinant rat galectin-1 (rabbit anti-galectin-l) (Cooper et al., 1991) was a gift from Drs. Sam Barondes, Hakon Leffler, and Doug Cooper (University of California, San Francisco). The mouse monoclonal antibody 9H10/4B10 (Pinol-Roma et al., 1988), directed against 51 hnRNP Al (M, ~34,000), was a generous gift of Dr. Gideon Dreyfuss (University of Pennsylvania) and is designated as mouse anti-hnRNP A1. The following mouse monoclonal antibodies were purchased from Transduction Laboratories (Lexington, KY): (a) mouse anti-Ran was used to detect the GTP-binding- protein Ran (M, ~ 25,000), involved in nucleocytoplasmic transport; and (b) mouse anti- karyopherin-B was used to detect the B-subunit (M, ~97,000) of the NLS receptor. Human autoimmune serum anti-Sm, used to detect Sm B (M, ~29,000) representing a core polypeptide of small nuclear ribonucleoprotein complexes (snRNPs), was purchased from The Binding Site (San Diego, CA). Human autoimmune serum used to detect the A polypeptide (M, ~33,000) specific to U1 snRNP, anti-U1 RNP, was also purchased from The Binding Site. Rabbit polyclonal antibody against phosphoserine was purchased from Zymed Laboratories (South San Francisco, CA). Wheat germ agglutinin (WGA) was purchased from Sigma (St. Louis, MO). Nucleotides corresponding to the monomethylated cap structures at the 5’-end of RNA polymerase II transcripts, m7GpppA and m7GpppG, were purchased from Boehringer Mannheim (Indianapolis, IN). The peptide NELALKLAGLDINKTG corresponds to residues 34-50 of PKI and has been shown to serve as the latter’s Nuclear Export Signal (NES), with the underlined leucine and isoleucine residues being critical for activity (Wen et al., 1995). This peptide was synthesized by the Macromolecular Structural Facility at Michigan State University and is hereafter designated the NES peptide. 52 The Nuclear Exmrt Assay in Digitonin-Perrneabilized Cells The following protocol is described for a typical assay (Fig. 1) derived from one tissue culture dish (28 cm2 surface area). Cells, at ~30% of confluent density, were washed with transport buffer (TB; 20 mM Hepes, pH 7.3, 110 mM KOAc, 2 mM EGTA, and 2 mM Mg(OAc)2) The cells were permeabilized in TB containing digitonin (1.75 11M) for 5 min. at 4 °C. After removal of the digitonin solution containing the soluble cytosolic components, 3 ml TB, supplemented with 1 mM dithiothreitol, 5 ug/ml aprotinin, 5 rig/ml pepstatin, and 10 ug/ml leupeptin, were added onto the permeabilized cells and incubated for various times of the export assay (Fig. 1). The supernatant was then collected as the Transported Fraction (TF). To 1.5 ml of TF, 15 1.11 deoxycholate (10%, w/w) and 150 [.11 trichloroacetic acid (100%) were added and the mixture was incubated at 4 °C for 30 min. The suspension was then centrifuged (12,000 x g; 15 min.) and the precipitate was washed with acetone. The precipitate was resuspended in 40 111 1% deoxycholate, 0.1 N NaOH; 10 111 of 5X SDS-PAGE sample buffer was then added and the entire sample was subjected to electrophoresis. In each experiment, the corresponding nuclear fraction (NF) was harvested from the same culture dish by solubilization in 50 ul of SDS-PAGE sample buffer. The components of the nuclear residue were analyzed by SDS-PAGE. 53 Figure 1: Schematic illustration of the digitonin-permeabilized cell system for the assay of nuclear export. Monolayer cultures of mouse 3T3 cells were treated with digitonin and then incubated with transport buffer. The presence of galectin-3 in the nuclear fraction was monitored by immunofluorescence and by Western blotting. The exported nuclear proteins were analyzed by Western blotting. 54 Soluble Digitonin cytosolic Components ‘~. I ~-‘---l” r- Transport Buffer At various times \ Exported 1 Nuclear \ ,’ components ’ ~_—’ Nuclear Fraction Transported Fraction 55 One- and Two—dimensional Gel Electrophoresis Proteins were resolved on one-dimensional gels, using 12.5% SDS-PAGE as described by Laemmli (1970). For two-dimensional gels, nonequilibrium pH gradient electrophoresis (NEPHGE) in the first dimension and SDS-PAGE in the second dimen- sion were used as described by O’Farrell et al. (1977) with the following minor modifi- cations. The NEPHGE gels were made using only pH 3-10 arnpholines and the electro- phoresis was can'ied out at 400 V for 5 h. Standard proteins for two-dimensional gels (BioRad, Richmond, CA) were used to track the pH gradient. Western Blotting and Quantitation For irnmunoblotting, proteins resolved by SDS-PAGE were transferred electrophoretically onto Immobilon-P membranes (Millipore, Bedford, MA) in buffer containing 25 mM Tris, 193 mM glycine, and 10% methanol. The membranes were blocked for several hours at room temperature with 10% non-fat dry milk in T-TBS (10 mM Tris, pH 7.5, 0.5 M NaCl, 0.05% Tween-20). After two brief washes with T-TBS, the membranes were incubated with primary antibody (rat anti-galectin-B, rabbit anti- galectin-1, mouse anti-hnRNP A1, mouse anti-Ran, mouse anti-karyopherin-B, human anti-U1 RNP and human anti-Sm) at room temperature for 1 h. The membranes were rinsed twice and then washed three times with T-TBS (one 15-min. wash and two 5-min washes). They were then incubated with the appropriate alkaline phosphatase-conj ugated 56 secondary antibody (goat anti-rat immunoglobulin, goat anti-rabbit immunoglobulin, goat anti-mouse immunoglobulin, or goat anti-human immunoglobulin, all at 1:2000 dilution) for 30 min. at room temperature. After rinsing twice and washing in T-TBS (one lS-min wash and two 5-min washes), the blots were developed using colorimetric substrates for alkaline phosphatase, nitroblue tetrazolium and 5-bromo-4-chloro-3-indolyl phosphate. For quantitation, the blots were scanned using a Hewlett-Packard ScanJet 3C and the DeskScan 11 program (Hewlett Packard, Palo Alto, CA). The image was then analyzed using Image Quant (Molecular Dynamics, Sunnyvale, CA) and the pixel values Of the individual bands were integrated to yield a value for intensity. Immunofluorescence For immunofluorescence microscopy, 3T3 cells were seeded onto glass coverslips placed in 6-well (8 cm2/well) cluster dishes. In the basic protocol, cells were washed twice with phosphate-buffered saline (PBS, 140 mM NaCl, 2.68 mM KCl, 10 mM Na,HPO,, 1.47 mM KH2P04, pH 7.4). and fixed for 20 min. at 4 °C with 4% paraforrnaldehyde in PBS. Cells were washed twice in PBS and then permeabilized with 0.2% Triton X-100 in PBS for 5 min. at 4 °C. After permeabilization, the cells were again washed twice with PBS and incubated with 0.2% gelatin in PBS for 1 h at room temperature. 57 Several variations of this basic protocol were also performed: (i) the cells were treated with digitonin as in the export assay (1.75 11M; 5 min.; 4 °C), followed immediately by fixation with paraformaldehyde and permeabilization with Triton X-100 as described above; (ii) the cells were treated with digitonin as in the export assay, fixed with paraformaldehyde but without the additional permeabilization with Triton X-100; and (iii) the cells were permeabilized with digitonin, incubated with TB for 30 min., followed by fixation with paraformaldehyde and permeabilization with Triton X-100 as described for the basic protocol. In all cases, the cells, after the “blocking” incubation in 0.2% gelatin in PBS, were washed in T-TBS. They were then incubated with rat anti-galectin-3 (250 ug/ml in T- TBS containing 0.2% gelatin) for l h at 4 °C. The cells were washed three times (15 min. each) with T-TBS and then incubated with fluorescein-labeled goat anti-rat irnmunoglobulin (10 ug/ml in T-TBS containing 0.2% gelatin) for 30 min. at room temperature. The coverslips were washed three times in T-TBS (15 min. each) and then mounted in Perma Fluor (Lipshaw Immunon, Pittsburgh, PA) on glass slides. Fluorescence staining was analyzed using a Meridian Instruments (Okemos, MI) Insight confocal laser scanning microscope. 58 RESULTS Exmrt of Galectin-3 from Nuclei of Digitonin-permeabilized cells The digitonin-penneabilized cell system, used to assay the import of protein bearing NLS (Adam et al., 1990; Moore and Blobel, 1993) was adapted to analyze the export of galectin-3 from the nucleus. The presence of galectin-3 in the nuclear residue was monitored by immunofluorescence and by Western blotting of NF. In parallel, TF, containing the exported nuclear components, was also analyzed for galectin-3 (Fig. 1). In accord with our previous observations (Laing and Wang, 1988; Wang et al., 1995), mouse 3T3 fibroblasts, fixed with paraformaldehyde and permeabilized with Triton X-100, yielded prominent nuclear and discernible cytoplasmic staining with anti- galectin-3 (Fig. 2A). When 3T3 cells were first treated with digitonin to permeabilize the plasma membrane, then fixed and treated with Triton X-100 to permeabilize the nuclear membrane, a similar staining pattern of the cell nucleus was observed (Fig. 2B). Cytoplasmic galectin-3 was lost upon permeabilization of the plasma membrane. The galectin-3 detected by this procedure (Fig. 2B) is inside the nucleus. This conclusion is based on the observation that digitonin-permeabilized cells, fixed with paraformaldehyde but without Triton X-100 permeabilization of the nuclear membrane to allow antibody accessibility, yielded no staining (Fig. 2C). When the digitonin-permeabilized cells were incubated in TB for 30 minutes prior to fixation and Triton X-100 permeabilization, galectin-3 staining was diminished drastically (compare Fig. 2D with Fig. 2B). These 59 .- .- Figure 2: Comparison of the immunofluorescence staining for galectin-3 in 3T3 cells before and after differential permeabilization. (A) Cells fixed with paraformaldehyde (4%; 20 min; 4 °C) and permeabilized with Triton X-100 (0.2%; 5 min; 4 °C); (B) Cells permeabilized with digitonin (1.75 11M; 5 min; 4 °C), followed immediately by fixation with paraformaldehyde and permeabilization with Triton X-100; (C) Cells permeabilized with digitonin and fixed with paraformaldehyde; and (D) Cells permeabilized with digitonin, incubated with transported buffer for 30 min, prior to fixation with paraformaldehyde and treatment with Triton X-100. Galectin-3 was detected with rat anti-galectin-3(250 ug/ml) and fluorescein-conjugated goat anti-rat immunoglobulin. 60 results suggest that nuclear galectin-3 can be exported during the 30 minute incubation in TB. This conclusion is corroborated by Western blot analysis of the galectin-3 polypeptide, both in the nuclear residue as well as in the soluble transported fraction. The level of galectin-3 in the nuclear residue is decreased considerably after the 30—minute incubation (Fig. 3, NF). Over the same time period, the level of galectin-3 in the supernatant rose correspondingly (Fig. 3, TF). Thus, both the immunofluorescence and immunoblotting analyses yielded consistent data, indicating that there was a rapid export of the galectin-3 polypeptide from the nuclei of digitonin-permeabilized 3T3 cells. This export assay has also been carried out using simian Cos-1 cells, with essentially the same results. Kinetics of Expgrt and Temmrature Demndence The rate of export of galectin-3 from digitonin-permeabilized 3T3 cells was temperature dependent. It was rapid at 37 °C; the half-life of galectin-3 in the nuclear residue was less than 5 minutes and less than 15% of the original nuclear galectin-3 remained after a 30-minute incubation in TB (Fig. 4). Parallel assays carried out at 25 °C and 0 °C showed that the rate of exit for galectin-3 decreased with lowering of the temperature. At 0 °C, for example, at least 40% of the galectin-3 remained with the nuclear residue over the 30-minute time course monitored. Because of the rapidity of the 61 Figure 3: Western blotting analysis for galectin-3 in the nuclear fraction (NF) and the transported fraction (T F ) before and after a 30-min. export assay. Galectin-3 was detected using rat anti-galectin-3 (250 ng/ml) and alkaline phosphatase-conjugated goat anti-rat immunoglobulin. The solid and dotted lines on the right highlight the positions of migration of the doublet corresponding to galectin-3 in NF at the beginning of the assay. The galectin-3 in TF consists of only the upper band of the doublet. The asterisk highlights the presence of a band in TF reactive with anti-galectin—3, most probably representing a degradation product. The numbers on the left indicate the positions of migration of molecular weight markers. 62 NF TF t(min) 0 30 0 30- 93- 67- 56" 42- 23— 17- 11- 63 t(min) o 2 4 8 16 30 Figure 4: Temperature dependence of the kinetics of galectin-3 export in the digitonin- permeabilized cell assay. The amount of galectin-3 remaining in the nucleus was monitored by Western blot analysis of NF at various times. The solid and dotted lines on the right highlight the positions of migration of the doublet corresponding to galectin-3. The arrowhead indicates the position of migration for Sm B, a nuclear protein that is not exported in the assay, and thus provides an indication that material from approximately the same number of nuclei were electrophoresed in each lane. 64 export system at 37 °C, all subsequent experiments used samples after an 8-minute incubation in TB. In these experiments, the export assay was carried out on cells incubated with fresh TB, after the removal of the digitonin solution used to permeabilize the plasma membrane of the cells. We have found that we can obtain essentially the same results by simply supplementing the digitonin solution with dithiothreitol and protease inhibitors, without the removal of the detergent. Thus, it appears that the continued presence of digitonin during the export assay does not extract or deplete factors affecting the transport itself. The Exit of Nuclear Galectin-3 is Selective In the course of these studies, careful analysis Of Western blots indicated that, consistently, galectin-3 is revealed as a doublet in the nuclear residue whereas only the upper band of the doublet can be found in the transported fraction (Fig. 3). We had previously Observed that, in 3T3 fibroblasts, galectin-3 can be found in two isoforms: (a) a phosphorylated form (pl ~8.2); and (b) a non-phosphorylated species (pl ~8.7). The former is present both in the nucleus and in the cytoplasm, while the latter is found exclusively in the nucleus (Cowles et al., 1990). On this basis, the nuclear residue and the transported fraction were subjected to two-dimensional gel electrophoresis, followed by Western blotting. The nuclear residue, which contained the galectin-3 doublet on 65 single-dimension SDS-PAGE, yielded two spots on the two-dimensional gels, with the upper band corresponding to the pl ~8.2 species and the lower band corresponding to the pI ~8.7 isoform (Fig. 5A). In contrast, the transported fraction, which contained only the upper band of galectin-3, yielded exclusively the pl ~8.2 spot (Fig. 5B). On this basis, we conclude that the upper band of the galectin-3 doublet represented the phosphorylated form while the lower band corresponded to the nonphosphorylated species. This conclusion is supported by two additional lines of evidence. First, when the nuclear residue is immunoblotted with anti-phosphoserine, only the upper band of the galectin-3 doublet showed reactivity (data not shown). This is consistent with the identification of Ser-6 as the major site of phosphorylation of the canine homolog of galectin-3 (Huflejt et al., 1993). Second, an export assay was carried out using 3T3 cells that had been metabolically labeled in the presence of 32P0,. The TF was subjected to lactose affinity chromatography and the bound material was resolved on SDS gels. The single galectin-3 band, identified by immunoblotting, was also radioactive upon autoradiography (data not shown). Although the exported galectin-3 consists of only the phosphorylated form, the majority of the galectin-3 molecules was found to be ultimately lost from the nuclear residue. Moreover, on the basis of the intensity of the immunoblotted bands, the amount of galectin-3 lost from the nucleus (Fig. 3, NF) can be approximately accounted for by the amount appearing in the transported fraction (Fig. 3, TF). Thus, it appears that the non-phosphorylated galectin-3 polypeptide (lower band) is first phosphorylated (to 66 A B NEPHGE {1 1‘3 * * 3.5 (I) if": - ".53” D (0 Basic Acidic Basic Acidic Figure 5: Two-dimensional gel electrophoretic analysis for galectin-3 in NF before the export assay (A) and in the TF after an 8-minute export assay (B). The samples were subjected to two-dimensional NEPHGE/SDS-PAGE and Western blotting. The asterisk marks the reference protein rabbit muscle glyceraldehyde 3-phosphate dehydrogenase, which yielded a pI value of 8.5. 67 become the upper band) prior to export. This notion is supported by a comparison of the ratios of the upper to lower bands at various time points during transport at 37 °C (Fig. 4). These results suggest that phosphorylation may be important for the exit of galectin-3 from the nucleus. The observation that only the phosphorylated form of galectin-3 appears to be exported suggests the system is highly selective in terms of substrates for export. Thus, we determined the export or retention of several nuclear proteins, on the basis of antibody reagents available. Karyopherin-B and Ran, two factors required for the import of NLS- bearing proteins, were also found to be exported. This conclusion can be derived by comparing the initial and 8-minute samples of either the nuclear residue (Fig. 6, lanes 1-2 and 5-6) or the exported material (Fig. 6, lanes 3-4 and 7-8). Galectin-l, belonging to the same lectin family as galectin-3, has also been identified as a nuclear splicing factor (Vyakamam et al., 1997). Like galectin-3, it is exported from the nucleus in our assay (Fig. 6, lanes 9-12). On the other hand, polypeptides of the snRNPs, Sm B representing the core polypeptide of several of the snRNPs and the A polypeptide specific to U1 snRNP, were not exported in our assay (Fig. 6, lanes 13-20). Finally, the hnRNP protein A1 is exported very slowly (relative to galectin-3), if at all (Fig. 6, lanes 21-24). Thus, the specificity of the galectin-3 transport system is manifested by: (a) comparing galectin- 3 with other components of the nuclear splicing machinery; and (b) comparing the isoelectric variants of the galectin-3 polypeptide itself. 68 1234 5678 9101112 1...... -- _——' O 13 14 15 16 17 18 19 20 21 22 23 24 - Figure 6: Comparison of several nuclear proteins in terms of their export or retention in the digitonin-permeabilized cell assay. Samples were subjected to Western blotting using the following antibodies: lanes 1-4, mouse anti-karyopherin-B (250 ng/ml); lanes 5-8, mouse anti-Ran (250 ng/ml); lanes 9-12, rabbit anti-galectin-l (1 :200 dilution); lanes 13-16, human anti-Sm (1: 10,000 dilution); lanes 17-20, human anti-U1 RNP (1: 10,000 dilution); and lanes 21-24, mouse anti-hnRNP A1 (1: 1000 dilution). The first two lanes of each set represent NF, at the beginning of the assay and after an 8-minute export period, and the last two lanes of each set represent the corresponding samples ofTF. 69 Inhibition of Galectin-3 Exmrt The plant lectin WGA, which binds N-acetyl-D-glucosamine (GlcNAc)- containing glycoconjugates, has been shown to inhibit the nuclear import of proteins (Finlay et al., 1987), as well as nuclear export of RNA (Yoneda et al., 1988; Bataille et al., 1990; Neuman de Vegvar and Dahlberg, 1990; Dargemont and Kuhn, 1992). The presence of WGA. in our assay inhibited the export of galectin-3 from the nucleus (Fig. 7). This was the case when WGA was added either to the permeabilization buffer containing digitonin or to TB. The effect of WGA was most pronounced when it was present during both permeaibilization and transport phases of the assay. Moreover, the effect of WGA can be reversed by its saccharide ligand, GlcNAc, but not by a non- binding saccharide, D-galactose (Gal) (Fig. 7). Similar results were obtained when the export of Ran was analyzed. Inasmuch as it has been shown that WGA has no effect on diffusion of fluorescent dextrans in and out of the nuclear pores (Jiang and Schindler, 1987; Wolff et al., 1988; Yoneda et al., 1988), these results further argue against the notion that the observed export of galectin-3 was due to leakage and diffusion. On the basis of our previous documentation that galectin-3 is required for the splicing of pre-mRNA (Dagher et al., 1995; Vykarnam et al., 1997) and that its detection in the nucleus is sensitive to ribonuclease (Laing and Wang, 1988; Hubert et al., 1995), we tested the effects of the nucleotides m7GpppG and m7GpppA. These correspond to the monomethylated, inverted guanosine cap structure, m7G(5’)ppp(5’)N, of RNA polymerase II transcripts and have been implicated to facilitate RNA export from the 70 _ _ WGA + WGA + WGA GlcNAc Gal t(min) O 8 8 8 8 GaleCtln‘3 ‘ nun—- - m“ é m- rams Ran Figure 7: The effect of wheat germ agglutinin(WGA)on the export of galectin-3 and Ran in the digitonin-permeabilized cell assay. Samples of NF, at the beginning of the assay and after an 8-minute export period, were subjected to Western blotting using antibodies against galectin-3 (rat anti-galectin-3, 250 ng/ml) and against Ran (mouse anti- Ran, 250 ng/ml). The concentration of WGA was 500 rig/ml and the concentrations of GlcNAc and Gal were 100 mM. 71 nucleus (Hamm and Mattaj, 1990). Although neither nucleotide, at a concentration of 2 mM, had an observable effect on galectin-3 export, we were surprised to find that m7GpppA (2 mM) in combination with the NES peptide (50 ug/ml) yielded partial inhibition (Fig. 8A). The sequence of the NES peptide corresponds to residues 34-50 of PKI, found to be responsible for the latter’s export fiom the nucleus, but the addition of NES peptide (50 rig/ml) by itself had little effect on transport in our assay (data not shown). Higher concentrations of m7GpppA (4 mM) and NES peptide (700 pig/ml) yielded greater (but still only partial) inhibition (Fig. 8B). This effect was not observed with the combination m7GpppG plus NES peptide (Fig. 8A and 8B). Moreover, the export of Ran was not similarly affected by the combination of m7GpppA plus NES peptide (Fig. 8B). These results suggest that at least some of the galectin-3 is exported in association with an RNP and that this RNP contains both the monomethylated cap structure m7GpppA and polypeptide(s) bearing NES-like sequences. This notion is schematically illustrated in Figure 9. 72 A_ 2mM 2mM mGpppG mGpppA - _ - 50 ug/ml _ 50 ug/ml . NES NES t(min) o 8 8 8 8 8 - “—— —— _ #— - Galectin-3 B_ 700 11ng NES t . _ _ 4mM 4mM (mm) mGpppG mGpppA - ‘15—" “‘v - Gal-3 - ~— -. . Ran Figure 8: The effects of 5'-cap structure nucleotide analogs and of a peptide bearing a nuclear export signal (NES) on the export of galectin-3 and Ran in the digitonin permeabilized cell assay. Samples of NF, at the beginning of the assay and after an 8- minute export period, were subjected to Western blotting using antibodies as described in the legend to Figure 7. In panel A, the nucleotides, m7GpppG and m7GpppA, were used at a concentration of 2 mM and the NES peptide was used at a concentration of 50 ug/ml. In panel B, the corresponding concentrations of nucleotides and peptide were 4 mM and 700 ug/ml, respectively. 73 Figure 9: Schematic diagram illustrating the association of galectin-3 with a ribonucleoprotein complex in TF of the export assay. Galectin-3 is represented by an oval (Gal3) containing a nuclear export signal (NES). The horizontal line depicts a mRN A, with a cap structure (mGpppA) at the 5'-end and a poly A tail (dotted line) at the 3'-end. The sphere (CBP) represents cap-binding protein(s). Other proteins associated with the RNA may also carry NES sequences. 74 DISCUSSION Since its initial development by Adam et al. (1990), the digitonin-permeabilized cell system has contributed much to our understanding of nuclear protein import. By selectively permeabilizing the plasma membrane, digitonin provides an in vitro system that appears to preserve much of the native architecture of the cell, allowing for the elucidation of the components and interactions involved in the transport of macromolecular substrates across the nuclear envelope. We have now taken advantage of this selective permeabilization system to analyze the export of galectin-3 from the cell nucleus. The key findings of our study include: (a) galectin-3 is rapidly and selectively exported from the nucleus; (b) although both phosphorylated and nonphosphorylated isoforms of galectin-3 are found in the nucleus, only phosphorylated galectin is identified in the exported fraction; and (c) while the Sm B and U1 A polypeptides of snRNPs are retained in the nuclear residue, galectin-l , Ran, and karyopherin-B are also exported in the same assay; ((1) the rate of export is decreased by cold temperature and by the addition of WGA; and (e) galectin-3 export is at least partially inhibited by the addition of cap-like m7GpppA nucleotide plus NES peptide. At least three types of nuclear export signals (NES) have now been identified: (i) short sequences enriched in hydrophobic residues, especially leucines (leucine-rich NES), as originally identified in PKI (F antozzi et al., 1994) and HIV Rev protein (Meyer and Malirn, 1994); (ii) a 38-amino acid region containing many glycine residues (designated 75 M9), identified on human hnRNP Al (Michael et al, 1995) and on its insect homolog, hrp 36 (Visa et al., 1996); and (iii) a KN S sequence, identified on hnRNP K that, like the M9 region, can function both for import and export (Michael et al, 1997). A sequence matching the leucine-rich NES, with distinct spacing of the leucine residues, can be found in galectin-3 homologs from various species. If this sequence indeed serves as an NES for galectin-3, then one might expect that a peptide corresponding to the NES of PKI would compete for the export machinery and inhibit the galectin transport. The addition of the NES peptide failed to affect galectin-3 export. Moreover, in transfection experiments using a FLAG epitope-tagged galectin-3 construct, we have carried out site- directed mutagenesis on the leucine residue correponding to Leu-44 of PKI. This leucine residue had been demonstrated by Wen et al. (1995) to be critical for the export activity of the NES. We found no difference, however, in the nuclear versus cytoplasmic distribution of the FLAG-tagged galectin-3 in cells transfected with the wild-type and the mutant constructs (Y.G. Tsay, unpublished results). One difficulty in interpreting such experimental results may be related to the complex nature of many of the molecular species that are exported fi'om the nucleus. It is generally believed that the RNA substrates for nuclear export are probably all transported as RNP complexes (see reviews by Izaurralde and Mattaj (1995), by Gerace (1995), and by Moore (1996)). Most of these RNPs contain multiple polypeptides so it seems possible that mutational knockout of the putative NES on galectin-3 will not alter its export properties as long as it remains associated with an RNP that carries other NES- bearing polypeptide(s) (see Fig. 9). Indeed, this model is supported by the observation 76 that a combination of NES peptide and nucleotide analogs recognized by cap-binding proteins are required to inhibit galectin-3 export (Fig. 9). Furthermore, this model may help account for the previous results that the monomethylated cap structure is a facilitating, but not a required, factor for mRN A export (Harnm and Mattaj, 1990; Jarrnolowski et al., 1994). The presence of multiple export signals on these RNPS probably circumvents the absolute requirement of a particular signal during nuclear export. Our present results need to be discussed in the context of two other studies that have used the digitonin-permeabilized cell system to study nuclear export. Moroianu and Blobel (1995) used digitonin-permeabilized buffalo rat liver cells to preload the nuclei with fluorescently-labeled human serum albumin covalently conjugated with the NLS of SV40 large T-antigen. These preloaded nuclei were then incubated to investigate the conditions for the export of the fluorescent substrate. They found that nuclear export in their system required Ran and GTP hydrolysis. In contrast, the export of galectin-3 in our system was not dependent on any exogenously added Ran or GTP. In fact, we found that Ran was also exported from the nuclei during the course of the transport assay, and , to the best of our knowledge, this represents the first demonstration on the nuclear export of Ran. Thus, it seems possible that depletion of Ran (and GTP) during the preloading of the fluorescently labeled transport substrate and subsequent washing steps may account for the apparent requirement for these factors, as observed by Moroianu and Blobel (1995). 77 More recently, Yang et al. (1997) have used digitonin-penneabilized GrH2 rat hepatoma cells, which express elevated levels of glucocorticoid receptors, to study the effect of hormone withdrawal on receptor dissociation from chromatin and a novel nuclear export process that is stimulated by sodium molybdate (Na2M004) and ATP. The effect of sodium molybdate can be mimicked by protein tyrosine phosphatase inhibitors, including sodium tungstate, sodium vanadate, and heparin; it is counteracted by inhibitors of tyrosine kinase such as genistein and tyrphostin AG126 (Yang et al., 1997). The stimulation of in vitro export by these compounds is not rmique to glucocorticoid receptors but can also be observed with heat shock proteins hsp70 and hsp90, as well as hnRNP A1. Several similarities and differences with our present results become immediately obvious. First, hnRNP A1 is exported very slowly, if at all, in our 3T3 cell assay. This was true even in the presence of ATP and sodium molybdate. It should be noted that hnRNP A1 is known to shuttle between the cytoplasm and the nucleus (Pit‘lol-Roma and Dreyfuss, 1993) and a nuclear export signal, designated M9, has been identified on the polypeptide (Michael et al., 1995). The fact that hnRNP A1 failed to be exported rapidly in our present assay argues strongly against non-specific leakage being responsible for the observed exit of nuclear components such as galectin-3.. This notion is supported by the observation that while the nucleus contains both phosphorylated and nonphosphorylated galectin-3, only the phosphorylated isofonn appears in TF. The export of galectin-3 is also blocked by WGA, similar to the latter’s inhibition of the transport of preloaded NLS- 78 human serum albumin (Moroianu and Blobel, 1995), and of the ATP- and molybdate- stimulated export of glucocorticoid receptors (Yang et al., 1997). Second, the export Of glucocoticoid receptors from the nuclei of GrH2 rat hepatoma cells required ATP, in addition to molybdate (Yang et al., 1997). On the other hand, the addition of either ATP or its nonhydrolyzable analog ATPyS had little or no effect on the export of galectin-3 in our assay. This apparent discrepancy on energy requirements may reflect the monitoring of different export substrates. ATP hydrolysis may be required for staging of the glucocorticoid receptors into a subnuclear compartment for export whereas galectin-3 transport may not need such preparation. In any case, neither the export of galectin-3 nor the export of glucocorticoid receptors appear to be sensitive to inhibition by GTPyS, suggesting that GTP hydrolysis was not required. On the other hand, Moroianu and Blobel (1995) had reported such a GTP dependence in the export of preloaded NLS-human serum albumin. Finally, in contrast to the stimulatory effect of molybdate, tungstate, and vanadate on glucocorticoid receptor export (Yang et al., 1997), we found no effect of these oxyanions on galectin-3 transport. More strikingly, we observed inhibition of the export of galectin-3 by vanadyl cations. The basis of the vanadyl versus vanadate difference may again be related to our idea that at least some of the galectin-3 is exported from nucleus in the form of an RNP (Fig. 9). Thus, the details of the vanadyl inhibition will be 79 presented in the accompanying manuscript, as a part of the analysis of the chemical components in the exported fraction (Tsay et al. , accompanying paper). CHAPTER III NUCLEAR EXPORT OF GALECTIN-3 IN MOUSE 3T3 FIBROBLASTS II. CHARACTERIZATION OF THE EXPORTED COMPONENTS Yeou-Guang Tsay, Nancy Y. Lin, Patricia G. Voss, and John L. Wang Department of Biochemistry Michigan State University East Lansing, MI 48823 80 81 SUMMARY The nuclear components collected in the transported fiaction of a nuclear export assay (see accompanying manuscript) was analyzed in terms of the polypeptide and RNA components. Gel filtration of the exported nuclear material and analysis for galectin-3 showed that the lectin can be found in at least two sets of high molecular weight complexes (~650 kD and ~60 kD). In the presence of lactose, both of these complexes are disrupted and galectin-3 chromatographs to a position corresponding to ~ 30 kD polypeptide. In contrast, the overall elution profile of galectin-1 in the transported fraction is not drastically altered by lactose. The polypeptide components of the high molecular weight complexes containing galectin-3 are revealed by affinity adsorption on a lactose-agarose column, specific elution by lactose, and gel electrophoresis. These polypeptides are not bound to a control cellobiose column. The transported fraction of the nuclear export assay also contain RNA. In the low molecular weight range, the RNA species include tRNA (~ 80 nucleotides) as well as RNAS of ~100, 300, and 650 nucleotides. High molecular weight RNAS, ranging from ~1 kb to 5 kb, include poly A” mRN A as revealed by hybridization with an oligo(dT) probe. Compounds containing vanadyl cations (V 02“), such as vanadyl ribonucleoside complex, inhibit the export of both galectin-3 and RNA in our assay. All of these results are consistent with the notion that galectin-3 is associated with a ribonucleoprotein complex during its export from the nucleus. 82 INTRODUCTION In the previous manuscript, we documented that, in digitonin-penneabilized 3T3 fibroblasts, galectin-3 as well as several other nuclear proteins, is rapidly and selectively exported from the nucleus (Tsay et al, 1997). Several key findings were made: (a) although both phosphorylated and nonphosphorylated isoforms of galectin-3 are found in the nucleus, only phosphorylated galectin-3 is identified in the exported fraction; (b) while Sm B and U1 A of snRNPs are retained in the nuclear residue, galectin-1, Ran, and karyopherin-B are also exported in the same assay; and (c) galectin-3 export is inhibited by the addition of cap-like m7GpppA nucleotide plus a peptide bearing the nuclear export signal (NES) of the inhibitor of CAMP-dependent protein kinase (PKI). On the basis of these observations, the possibility was raised that galectin-3 may be exported in association with a ribonucleoprotein complex (RNP), with many polypeptides bound directly or indirectly with an RNA species. One major advantage of the digitonin-permeabilized export assay lies in the fact that the exported material can be collected in a soluble form, amenable for fiactionation and biochemical analysis. We have thus carried out preliminary physico-chemical characterization of the components of the exported nuclear components, with two main goals: (a) to obtain additional evidence that nuclear galectin-3 is associated with a high molecular weight complex; and (b) to delineate the number and, if possible, the identity 83 of the polypeptide and RNA species of the galectin-3 complex. The results of these studies are documented in the present communication. 84 EXPERIMENTAL PROCEDURES Cell culture and reagents NIH mouse 3T3 fibroblasts were purchased form the American Type Culture Collection (Rockville, MD). The cells were grown in Dulbecco's Modified Eagle Medium containing 10% calf serum, 100 U/ml penicillin, and 100 ug/ml streptomycin at 37°C in a humidified atmosphere of 10% C02. Generally, cells were seeded at a density of~1.5 x 10‘ cells/cm’, about 30% of the saturation density. Metabolic labeling with 32P- labeled orthophsophate was carried out at the concentration of 50 uCi/ml culture medium. Anti-Galectin-3 antibody is a rat monoclonal antibody developed against the Mac- 2 antigen (Ho and Springer, 1982); anti-galectin-l antibody is a polyclonal rabbit antiserum raised against rat galectin-1 (a generous gift from Dr. Barondes, University of Califomia, San Francisco); anti-Ran antibody is a mouse monoclonal antibody, purchased fiom Transduction Laboratories (Lexington, KY). Vanadyl sulfate hydrate was fiom Aldrich (Milwaukee, WI), vanadyl ribonucleoside complex (V RC) was from GIBO-BRL (Gaithersburg, MD), and ribonucleosides were from Boehringer-Mannheim (Indianapolis, IN). Agents purchased from Sigma (St. Louis, MO) include sodium metavanadate (NaVO3), tungstic acid (HZW04), sodium molybdate (NaQM004), and all of the secondary antibodies used for immunofluorescence and immunoblotting analyses. 85 Nuclear exmrt assay The details of the assay are describe in the accompanying manuscript (Tsay et a1. , 1997). For transported fraction (TF) used in gel filtration chromatography and immunoprecipitation analysis, VRC was added at a concentration of 10 mM immediately following, the collection of TF. TF used for gel filtration and immunoprecipitation was concentrated approximately 200-fold in Ultrafree-l 5 centrifugal filter device (Millipore, Bedford, MA) centrifuged at 3,000 x g. For immunofluorescence experiments, cells seeded on coverslips were incubated with 40 ug/ml digitonin at 0°C for 5 min. Reagents to be tested for stimulatory or inhibitory effects then added without removal of the digitonin solution. The cells were incubated at 25 °C for 8 min. To stop the reaction, cells were incubated in 3.7% of formaldehyde for 20 min at 0 °C. Fixed cells were washed in phosphate-buffered saline (PBS) twice for 10 min. Penneabilization with 0.2% Triton X-100 was carried out at room temperature for 5 min. After two 10—min washes, 0.2% of gelatin was added to the cells. Staining with antibodies was described in the accompanying manuscript (Tsay et al., 1997). Concentrations for primary antibodies are 2.5 jig/ml for anti-Ran and 25 ng/Inl for anti-galectin-3. FITC-conjugated anti-mouse IgG and anti-rat IgG antibodies were both used at 1:250 dilution. For immunoblotting experiments, cell were treated as described above. After the incubation at 25°C, the cell residue, designated as the nuclear fiaction (NF), was 86 collected in SDS-PAGE sample buffer. The sample was sonicated for 1 min at room temperature and heated at 100 °C before loading to a 12.5% polyacrylamide gel. The polypeptides resolved on polyacrylamide gels were electrotransferred onto Immobilon-P (Millipore, Bedford, MD) and immunoblotted with the indicated antibodies. Concentrations of primary antibodies for immunoblotting are 250 ng/ml for anti-Ran, 250 ng/ml for anti-galectin-3 and 1:1000 for anti-galectin-l. All of the secondary antibodies were used at 1:2000 dilution. Gel filtration chromatoggphy About 24 ml of Sephacryl S-300 HR (Phararnacia, Piscataway, NJ) were packed in a 1 x 30 cm Econo-Column column (Bio-Rad, Hercules, CA) according to the manufacturer's guide. The column is rinsed with transport buffer (TB) at the rate of 24 ml/hour and then equilibrated with the same buffer at the rate of 8 nil/hour before application of the sample. Molecular weight standards (100 ug each) include thyroglobulin (670 kDa), alcohol dehydrogenase (150 kDa) and carbonic anhydrase (29 kDa). Eluted components were collected in 13-drop (~ 0.75 ml) fractions. Protein concentration in each fraction was determined by the Bradford method (Bradford, 1976). Typically, TF derived from 4 x 10‘ cells, in a volume of 200 pl, was loaded onto the column and then eluted with TB. Polypeptide components in each fraction were precipitated in the presence of 10% trichloroacetic acid and 1% deoxycholate at 4°C for 30 min. The pellet was collected after centrifirgation at 12,000 x g for 15 min. After 87 rinsing with acetone, the pellet was resuspended with 0.1% deoxycholate-0.1 M NaOH and mixed with 5X SDS sample buffer. The samples were analyzed by SDS-PAGE and immunoblotting. Affini adso tion chromato h For saccharide adsorption experiments, 200 pl of concentrated TF were incubated with 40 ul of 1:1 slurry of lactose- or cellobiose-agarose (Sigma) overnight at 4°C. These sample was centrifuged at 12,000 x g for 10 sec, and the supernatant (unbound fiaction) was removed fi'om the beads. The agarose beads were then washed four times in TB containing 1% NP-40, 0.5% deoxycholate and 0.1% SDS. After the last wash, bound materials were eluted with 0.2 M lactose in TB and designated as eluted fiaction. The eluted fraction was resolved on a 12.5% polyacrylamide gel and analyzed by silver staining. For antibody adsorption experiments, concentrated TF was incubated with 3 ug of anti-galectin-3 overnight at 4°C. Twenty microliters of 1 :1 protein G-agarose were added to the solution and mixed with rocking at room temperature for 2 hours. The sample was centrifirged at 12,000 x g for 10 sec, and the supernatant (unbound fraction) was removed and subjected to RNA analysis. Analysis of RNA commnents To samples containing 32P-labeled RNAS were added 0.1% NP40, 0.1% SDS, and 10 ug/ml tRNA and then treated with 10 jig/ml of proteinase K at 37 °C for 10 min. 88 RNAS were then isolated by standard phenol extraction and ethanol precipitation procedures (Sarnbrook et al., 1989). RNA pellets were resuspended in formaldehyde loading buffer and resolved on a fonnaldehyde-l .2% agarose gel or an 8% polyacylamide gel. RNAS were then visualized by autoradiography. For detection of poly(A)" RNAS, unlabeled RNAS were prepared as described and then resolved on a formaldehyde-8% agarose gel. RNAS were hybridized in dried gels with 32P-labeled oligo(dT),2_,8. (GIBCO-BRL, Gaithersburg, MD) at room temperature using the standard procedure (Sambrook et al., 1989). RNAS hybridized with oligo(dT) were visualized by autoradiography. 89 RESULTS Gel Filtration of Exmrted Nuclear Compgnents and Analysis for Galectin-3 In the previous manuscript, we documented the use of digitonin-penneabilized 3T3 fibroblasts to analyze the export of galectin-3 fi'om the nucleus (Tsay, accompanying manuscript). We defined the material remaining in the nuclear residue alter a given period of incubation as the Nuclear Fraction (N F). The exported material is collected in the Transported Fraction (TF). One key observation was that the export of galectin-3 can be inhibited, at least partially, by the simultaneous addition of the cap-like nucleotide m7GpppA plus a peptide bearing the NES from PKI. The possibility was raised that galectin-3 may be exported in association with an RNP, which contain signals that facilitate nuclear export in both the 5'-cap structure of the RNA species as well as peptide sequences of component polypeptide(s). Because one major advantage of the digitonin- penneabilized export assay lies in the fact that the exported material can be collected in a soluble form amenable for fractionation and biochemical analysis, we have carried out preliminary physical and chemical characterization of the components of TF. First, TF was concentrated approximately ZOO-fold and subjected to gel filtration on a column of Sepharcryl S-300 HR. Individual fiactions eluting from the column were subjected to SDS-PAGE and Western blotting for galectin-3. The elution profile for galectin-3 in TF showed three zones, with peaks centered around: (a) ~650 kD; (b) ~60 90 kD; and (c) ~15 kD (Fig. 1A). Previous gel filtration analyses of galectin-3, purified from 3T3 cells (Roff and Wang, 1983) and from E. coli expression system (Hsu et al., 1992), showed that the protein chromatographed as a ~30 kD species, corresponding to the monomeric polypeptide. On this basis, we can immediately conclude that galectin-3 in TF existed in at least two sets of complexes: (a) the ~650 kD group; and (b) the ~60 kD group. The presence of galectin-3 in the zone centered around ~15 kD may complicate the molecular weight estimates because it suggests that either galectin-3 or an associated component may interact with the Sephacryl backbone and get retarded in the chromatographic column. We have carried out similar gel filtration studies using a number of other matrices (e. g. Sepharose, Sephadex); the same overall conclusion, that galectin-3 in TF was associated with high molecular weight complexes, was obtained. The individual fiactions from the Sepharcryl S-300 HR column were also subjected to Western blotting for galectin-1. There was a broad zone of elution, with a peak slightly higher in molecular weight and another one just slightly lower in molecular weight than the 29 kD marker (Fig. 1B). This result is consistent with the report of Cho and Cummings (1995), who showed that galectin-1, on gel filtration, yielded two poorly resolved peaks corresponding to the monomeric polypeptide (~15 kD) and a dimer (~30 kD). The ratio of monomer to dimer depended on the concentration of the protein in solution. Because the members of the galectin family exhibit very similar carbohydrate- binding specificities, the fact that galectin-l chromatographed at the expected position suggests that the retardation of galectin-3 in the ~15 kD zone (Fig. 1A) is most probably not due to protein interactions with the carbohydrate backbone of the Sephacryl matrix. 91 670K 150K 29K . V V v A ’ ‘ I --.—.- - -— ——---- -'“" B .2...- . Figure 1: Gel filtration of the exported nuclear components in TF and analysis for galectins-l and -3. The column (1 .3 x 29 cm) was packed with Sephacryl S-300 HR and was equilibrated with TB (containing supplements?) HOW MUCH TF LOADED? IN WHAT VOLUME? Fractions of 0.5 ml were collected, subjected to SDS-PAGE and immunoblotting with anti-galectin-3 (panel A) or anti-galectin-I (panel B). The numbers at the top indicate the positions of migration of molecular weight markers: thyroglobulin (670 K), alcohol dehydrogenase (150 K), and carbonic anhydrase (29 K). 92 Using purified galectins-l and -3 , previous studies had shown that the disaccharide ligand lactose has little or no effect on the chromatographic behavior of these lectins (Roff and Wang, 1993; Hsu et al., 1992; and Cho and Cummings, 1995). The inclusion of Lac in the chromatographic medium had little or no effect on the overall position of elution for galectin-1 in TF (Fig. 2). There appeared to be some diminution of the peak >29 kD and accentuation of the peak < 29 kD. One possibility is that Lac shifted the monomer-dimer equilibrium in favor of subunit dissociation. An alternative possibility is that the > 29 kD peak represents some association of galectin-1 with another component and that this complex is disrupted by Lac binding to galectin-1. For galectin-3 in TF, however, the inclusion of Lac in the chromatographic medium drastically altered its elution profile (Fig. 2). The peak at ~650 kD zone disappeared. The peak at ~60 kD zone was also diminished. The majority of the galectin-3 chromatographed to a position slightly lower in molecular weight than the 29 kD marker. These results suggest that the high molecular weight complexes with which galectin-3 is associated in TF is sensitive to Lac-binding. Individual fractions in the ~650 kD zone derived from colmnns carried out in the absence and presence of Lac were compared by SDS-PAGE and silver staining. The polypeptide compositions of these fractions fi'om the column with Lac were quite distinct from the corresponding fractions fi'om columns without the disaccharide (data not shown). These results are consistent with the notion that Lac removed not only galectin-3 from the ~650 kD zone but also other polypeptides in the high molecular weight complex. Finally, it should be noted 93 670 K 150 K 29 K '---—-- -__.---—- .. -41.-- 1—4' + Galectin-3 ..._. a! ’ - “a... d + Galectin-1 Figure 2: The effect of Lac on the gel filtration profile of galectins-1 and -3 in TF. The column and the conditions of chromatography are as described in the legend to Figure 1. In experiments testing the effect of Lac, the column was equilibrated and developed with TF containing 0.2 M Lac. 94 that, even in the presence of Lac, traces of galectin-3 could still be observed over a broad region of the column, ranging in molecular weight from <600 kD to ~60 kD (Fig. 2). Affinig Chromatography of Transpgrted Fraction on a Lactose Column The exported material in TF was also subjected to affinity adsorption on Lac- agarose beads. After extensive washing, the bound material was eluted with Lac. This material revealed a very complex composition of polypeptides upon SDS-PAGE and silver staining (Fig. 3). The most prominent bands corresponded to polypeptides of : (i) ~15 kD; (ii) ~30 kD; (iii) ~40 kD; (iv) ~50 kD; (v) ~58 kD; and (vi) ~93 kD. With the exception of the ~50 kD band, none of these prominent bands can be found when TF was subjected to a parallel adsorption on cellobiose-agarose beads (Fig. 3). On the basis of the Lac-binding property of the galectin family members and on the basis of immunoblotting, galectins-l and -3 can be identified. The other bands most probably represent polypeptides of a complex that have bound to the Lac beads through their association with either galectins-l or -3. The complexity of the polypeptide profile precludes a detailed comparison. However, it should be noted that the pattern presented in Figure 3 (Lac affinity adsorption) is very similar to: (a) that obtained from fractions of the ~650 kD zone of the Sephacryl S-300 HR column; and (b) that observed when nucleoplasm Of 3T3 fibroblasts is fractionated on an N-(e-aminocaproyl)-D- galactosarnine-Sepharose column and eluted with galactose (Laing and Wang, 1988). 95 Figure 3: Affinity adsorption of the polypeptide components in TF on Lac- agarose and cellobiose-agarose. Parallel samples of 200 111 were incubated with 40 pl of Lac-agarose or cellobiose-agarose. The material bound to the beads were eluted with TB containing 0.2 M Lac, subjected to SDS-PAGE and silver staining. The numbers at the left indicate the positions of migration of molecular weight markers. 96 221— 133.. 93— 67— 56— 42- 28— 23- ' 17— 11- (kDa) 97 The gel filtration studies on TF in the presence and absence of lactose suggested that the binding of the disaccharide disrupted the high molecular weight complexes containing galectin-3 (Fig. 2). Therefore, the question may be raised as to how galectin- 3-associated polypeptides can still be isolated on the basis of Lac-affinity adsorption (Fig. 3). One possible explanation may lie in our previous parenthetical note that, even in the presence of Lac, traces of galectin-3 could still be Observed over a broad range of the gel filtration column, from <600 kD to ~60 kD (Fig. 2). This may reflect dynamic and continuous reequilibration of galectin-3 with the high molecular weight complex and it is on this basis that the Lac affinity beads can trap some of the associated polypeptides. It may also be worthy to note that the efficiency of galectin-3 isolation (amount of galectin- 3 in the bound and Lac-eluted fiaction as a percent of galectin-3 in the unfractionated material) fi'om TF was much lower than the efficiency of adsorption of purified recombinant galectin-3 onto the same Lac-agarose matrix. One possible implication of this observation is that the affinity of galectin-3 for its saccharide ligand is decreased in complex formation. Alternatively, galectin-3 in TF may already be occupied with carbohydrate ligands with which Lac-agarose must compete for binding. Effect of Vanadyl Cations on the Exmrt of Galectin-3 The inhibition of galectin-3 export by the combination of 5’ cap-like nucleotide and NES peptide had suggested the possibility that the lectin was co-exported with RNA(s) (Tsay et al., 1997). The present finding that a significant portion of the exported galectin-3 can be found in a high molecular weight complex lends support to the 98 hypothesis. On the basis of previous reports documenting that VRC can stabilize RNA species from nuclease degradation (Berger and Birkenmeier, 197 9), we tested the effect of VRC on the preservation of the galectin-3 high molecular weight complex in TF. To our surprise, we found that VRC (10 mM) was a potent inhibitor of nuclear export in our assay (Fig. 4A). The active ingredient appears to be vanadyl (V O”) cations, inasmuch as vanadyl sulfate (10 mM) also inhibited nuclear export but little or no inhibition was Observed with a mixture of the four ribonucleosides (10 mM). The oxoanion of vanadium, vanadate (V 03'), did not Show inhibition at the same concentration. Nor did other transition metal oxoanions, molybdate and tungstate (data not shown). A comparison of the dose-response curves of inhibition showed that VRC was potent at concentrations as low as 5 mM (Fig. 48) while strong inhibition was observed only with higher concentrations (10 mM or greater) of vanadyl sulfate (Fig. 4C). The conclusion that VRC was more effective than vanadyl sulfate in suppressing nuclear export can also be derived from immunofluorescence analysis for galectin-3 in the nuclear residue. The level of galectin-3 appeared to be higher in the nuclear residue of VRC (10 mM)-treated cells (Fig. 5D) than the corresponding cells treated with vanadyl sulfate (10 mM) (Fig. 5E). Surprisingly, the simultaneous application of VRC and vanadyl sulfate (10 mM each) resulted in even greater inhibition than either reagent alone (Fig. 5F). This suggests the possibility that the effects of the two reagents may be additive and thus may be acting via different pathways or different populations of molecules. Parenthetically, the inhibitory effect of vanadyl cations is more potent that 99 Figure 4: The effect of vanadyl cations (V01) on the nuclear export of galectin-3, assayed by immunoblotting NF. In panels A-C, the first left hand lane represents NF at the beginning of the export assay. All the other lanes in each panel represent NF at the end of an 8-minute export period. Panel A: -, no addition; VRC, vanadyl ribonucleoside complex (10 mM); V0”, vanadyl sulfate (10 mM); Ribonucleoside, mixture of four ribonucleosides (2.5 mM each). Panels B and C: dose-response curves for VRC and Vanadyl sulfate, respectively. Figure 5: The effect of vanadyl cations (VO“) on the nuclear export of galectin-3, assayed by immunofluorescence analysis of NF. Galectin-3 was detected using rat anti- galectin-3 (250 ng.ml) and fluorescein-conjugated goat anti-rat immunoglobulin. A: NF at the beginning of the export assay. B-F: NF at the end of an 8-minute export period. B: no addition; C: wheat germ agglutinin (500 ug/ml); D: VRC (10 mM); E: vanadyl sulfate (10 mM); and F: VRC and vanadyl sulfate (10 mM each). 101 that of wheat germ agglutinin (Fig. 5C), a conclusion also corroborated by subjecting NF to Western blot analyses. Finally, the inhibitory effect of vanadyl cations on nuclear export is not restricted to galectin-3; a similar set of results was also obtained by analyzing for Ran. Immunofluorescence analysis of NF at the beginning of the export assay (Fig. 6A) and after an 8-minute export period (Fig. 6B) showed that Ran was rapidly exported. This is in agreement with the immunoblotting data presented in the accompanying manuscript (Tsay et al., 1997). To the best of our knowledge, the immunoblotting and immunofluorescence data together represent the first rigorous documentation that Ran is exported from the nucleus. Like that for galectin-3, the export of Ran was sensitive to inhibition by wheat germ agglutinin (Fig. 6C), by VRC (Fig. 6 D), and by vanadyl sulfate (Fig. 6E), with the strongest effect observed with a combination of VRC and vanadyl sulfate (Fig. 6F). Analysis of RNA Commnents in the Transpgrted Fraction An export assay was carried out using 3T3 cells that had been metabolically labeled with [32P]orthophosphate. The RNA components in TF collected from this assay were extracted, precipitated, and resolved on separate polyacrylamide and agarose gels. Autoradiography of the polyacrylamide gels revealed low molecular weight RNAS, with bands corresponding to: (i) ~80 nucleotides; (ii) ~100 nucleotides; (iii) ~300 nucleotides; (iv) ~650 nucleotides; (v) ~800 nucleotides; and (vi) >1000 nucleotides (Fig. 7A). With 102 Figure 6: The effect of vandyl cations NO“) on the nuclear export of Ran, assayed by immunofluorescence analysis of NF. Ran was detected using mouse anti-Ran (250 ng/ml) and fluorescein-conjugated goat anti-mouse immunoglobulin. A: NF at the beginning of the export assay. B-F: NF at the end of an 8-rninute export period. B: no addition; C: wheat germ agglutinin (500 ug/ml); D: VRC (10 mM); E: vanadyl sulfate (10 mM); and F: VRC and vanadyl sulfate (10 mM each). 103 Figure 7: Gel electrophoretic analysis of the RNA components in TE when the export assay is carried out in the absence and presence of vanadyl sulfate. The export assay was carried out using 3T3 cells metabolically labeled with [32P]orthophosphate and the RNA components of TF were isolated and resolved on an 8% polyacrylamide gel (panel A) or a 1.2% agarose gel (panel B). The radioactive RNA components were revealed by autoradiography. The numbers on the left indicate positions of migration of size markers in nucleotides (panel A) or in kb (panel B). In experiments testing the effect of vanadyl cations, the export assay was carried out in the presence of 10 mM vanadyl sulfate. 1.35- 0.87- 0.60- 0.31- 0.23- 0.19- 0.12- 0.07- (kb) VO2+ 104 4.9- 3.5- 2.0- 1.6- . 1.1- 0.6- (kb) 105 the exception of tRNA (75-85 nucleotides), the identities of the other small RNAS are not known. Autoradiography of the agarose gel revealed a smear ranging from 0.7 kb to ~5 kb, with prominent bands at ~ 1.2 kb and ~ 3 kb (Fig. 7B). An Oligo dT probe was used to hybridize to the high molecular weight RNA; on this basis, RNA species ranging fiom 1 kb to ~5kb can be identified as poly(A)+ mRN A (data not shown). Therefore, it appears that both low molecular weight RNA, as well as mRNA, are exported in our assay and can be collected in TF. We have made several attempts to delineate the RNA species associated with the high molecular weight galectin-3 complex, using either Lac affinity adsorption or immunoprecipitation. Due to technical difficulties that we have yet overcome, however, we have been unsuccessful in isolating and identifying the RNA components of this complex. By comparing the level of radioactivity and the RNA species in the u_nbgu_n_d_ fractions the Lac-beads or anti-galectin-3-beads with the corresponding unbound fractions of control beads (e. g. cellobiose-beads), we have ascertained that radioactive RNA is indeed bound to those beads that should trap galectin-3. Thus, what remains is the delineation of the number and identity of the RNA species. Just as we had demonstrated for the export of galectin-3 (and Ran) in terms of polypeptides, vanadyl cations also inhibited the export of RNA in our assay. The transport of both low molecular weight RNA species (Fig. 7A), as well as high molecular weight mRNA (Fig. 73), were suppressed. 106 DISCUSSION Taking advantage of the fact that the material in TF of the digitonin-permeabilized nuclear export assay can be collected in a soluble form, we have carried out initial characterization of the components of TF. The key findings of the present study include: (a) gel filtration of TF and analysis for galectin-3 showed that the lectin is associated with at least two high molecular weight complexes (~650 kD and ~60 kD); (b) both complexes are sensitive to disruption by the presence of the galectin ligand, Lac; (c) the polypeptide composition of the high molecular weight galectin-3 complex appears to be very complicated, with prominent bands of about 15 kD, 30 kD, 40 kD, 50 kD, 58 kD, and 90 kD; (d) TF also contains RNA, including small nuclear RNAS, as well as poly A+ mRNA; and (e) vanadyl cations (V 02“) inhibit the export of galectin-3 and the RNAS. These results provide new levels of information to two important previous observations. First, galectin-3 has been identified as a required factor in the splicing of pre-mRNA, assayed in a cell-free system (Dagher et al., 1995; Vyakamam et al, 1997). Saccharides such as Lac that bind to galectin-3 with high affinity inhibit the splicing reaction. The present finding that the high molecular weight galectin-3 complex is disrupted by Lac may provide a mechanism for the effect of the saccharide in the splicing reaction. In both the inhibition of splicing and in the disruption of the high molecular weight galectin-3 complex, it still remains to be determined whether Lac is competing 107 with a glycoconjugate ligand for the carbohydrate-binding site of galectin-3 or it is inducing a conformational change upon binding to the galectin-3 polypeptide. Using differential scanning calorimetry, it has in fact been demonstrated that Lac binding to galectin-3 induces a conformation change in the polypeptide (Agrwal et al., 1993). Coupled with our failure, despite various attempts, to identify a glycoconjugate ligand for galectin-3 in the nuclear fraction, we tend to favor the notion that a conformational change in the galectin polypeptide upon Lac binding dissociates the protein from its complex. This may, in turn, lead to loss of splicing activity. Second, we had documented, in the accompanying manuscript, that the nuclear export of galectin-3 was sensitive to inhibition by a combination of a nucleotide mimicking the 5’ cap structure of mRNAs and of a peptide bearing the NES from PKI. Coupled with its required role in the splicing reaction, the results suggested that galectin- 3 may be exported in the form of an RNP, with many polypeptides bound directly or indirectly with an RNA species. Thus, signals that facilitate nuclear export in both the 5’ cap structure of the RNA species and the peptide sequences of the component polypeptides needed to be neutralized before inhibition of export could be observed. Indeed, we found in the present study that gel filtration of the exported material yielded galectin-3 in at least two sets of high molecular weight complexes. It should be noted that although Lac disrupts the galectin-3 complex(es), the saccharide actually has little or no effect on the rate of the protein’s export from the nucleus (Y.G. Tsay, unpublished observations). Even in the presence of Lac, however, traces of galectin-3 could be observed over a broad range of the gel filtration column (from <600 kD to ~60 kD) and 108 these putative complexes may continue to serve the role of providing multiple signals for nuclear export. Although we have obtained some information on the complexity of the polypeptide composition of the galectin-3-associated particle and although we can document that TF contains RNA, we have yet to delineate the number and identity of the RNA species. If galectin-3 is indeed exported in the form of an RNP, then it would join two other pre-mRNA splicing factors that accompany their RNA substrates out of the nucleus. Both hnRNP A1 (Mayeda et al., 1992; Caceres et al., 1994) and the cap-binding complex that recognize the monomethylated guanosine cap structures at the 5’-end of RNA polymerase II transcripts (Izaurralde et al., 1994; Lewis et al., 1996) travel out of the nucleus with mRNAs (Visa et al., 1996a and Visa et al., 1996b). Once out of the nucleus, whether the RNA stays with galectin-3, as is with hnRNP A1 (Visa et al., 1996b), or the splicing factor is dissociated from the RNP, as is with the cap-binding complex (Visa et al., 1996a), are issues that need to be studied. In the course of the present study, we found that vanadyl cations strongly inhibited the nuclear export of both galectin-3 and RNAS. However, no effect could be demonstrated with vanadate, molybdate, or tungstate. This is to be contrasted to the stimulatory effect of these oxyanions on the nuclear export of the glucocorticoid receptor (Yang et al., 1997). Vanadium exhibits complex chemistry, fluctuating between vanadyl (oxidation state +4) and vanadate (oxidation state +5), with possible reduction-oxidation interconversions (Macara et al., 1980; Li et al., 1996). Over the short time course of our 109 export assays, however, the inhibitory effect on galectin-3 export appears to be restricted to compounds that give rise to vanadyl cations (V RC, vanadyl sulfate) whereas no such effects were observed with vanadates. This is consistent with recent observations that while both vanadyl cations and vanadate oxyanions can target protein phosphotyrosine phosphatases, distinct vanadate-independent and vanadyl-dependent pathways can be dissected (Li et al., 1996). Beside protein phosphotyrosine phosphatases, another set of targets susceptible to inhibition by ribonucleoside-vanadyl complexes are nucleases (Berger and Birkenmeier, 1979). Thus, it seems possible that VRC indirectly affects galectin-3 export through inhibition of a ribonuclease(s). This enzyme may have specifically digested an RNA that anchors galectin-3 -containing RNPS to the nuclear structure. Galectin-3 export is not allowed, when the anchor remains intact due the inhibition of the nuclease. This notion is consistent with our previous observation that association of galectin-3 with nuclear structure was sensitive to treatment with ribonuclease (Laing and Wang, 1988; Wang et al., 1995). Since VRC has been shown to have similar inhibitory effects on nuclear export of Ran and galectin-l, if this peculiar mechanism does exist, it may also regulate the export of Ran and galectin-1. So far, crucial evidence is still lacking and this possibility awaits firrther investigation. 110 CONCLUSIONS In this manuscript, we demonstrate that galectin-3 has nuclear shuttling activity; i.e., it can move between the nucleus and cytoplasm bidirectionally. The . nucleocytoplasmic distribution of galectin-3 is , in fact, a net result of counteraction between nuclear import and export. When nuclear import is predominant, this will lead to nuclear localization. In contrast, when nuclear export dominates over import, cytoplasmic localization will be seen. As a pre-mRN A splicing factor, galectin-3 shows differentiation localization as a function of proliferative states of cells, which distinguishes galectin-3 from other splicing factors. The major implication is that the splicing process may be modulated by nuclear accumulation of galectin-3. In conjunction with the fact that galectin-3 can shuttle between the nucleus and cytoplasm, it is possible that nuclear import may serve as a mechanism that upgrades RNA splicing activities, and nuclear export is a mechanism that down-regulates RNA splicing activities. On the other hand, we also report that galectin-3 appears to be exported in complexes that contain RNAS capped with mGpppA dinucleotide and polypeptides bearing NES signals. This finding appears to suggest that galectin-3, along with other components in these complexes, may participate in RNA metabolism or other cellular processes subsequently. In other words, nuclear export of galectin-3 is not only a way to 111 turn ofir nuclear activities, but also a mechanism to turn on certain cytoplasmic activities. It will be interesting to study the fate of these ribonucleoprotein complexes in cytoplasm as well to explore the roles of galectin-3 in the process. LIST OF REFERENCES 112 LIST OF REFERENCES Ackerman, S. J., S. E. Corrette, H. F. Rosenberg, J. C. Bennett, D. M. Mastrianni, A. Nicholson Weller, P. F. Weller, D. T. Chin, and D. G. Tenen. 1993. Molecular cloning and characterization of human eosinophil Charcot-Leyden crystal protein (lysophospholipase). Similarities to IgE binding proteins and the S-type animal lectin superfamily. J. Immunol. 150:456-468. Adam, S. A., R. S. Marr, and L. Gerace. 1990. Nuclear protein import in permeabilized mammalian cells requires soluble cytoplasmic factors. J. Cell Biol. 111:807-816. Agrwal, N., Q. Sun, S.-Y. Wang, and J. L. Wang. 1993. Carbohydrate-binding protein 35: I. Properties of the recombinant polypeptide and the individuality of the domains. J. Biol. Chem. 268:14932-14939. Ahmed, H. and G. R. Vasta. 1994. Galectins: conservation of functionally and structurally relevant amino acid residues defines two types of carbohydrate recognition domains. Glycobiology 4: 545- 549. Aitchison, J. D., G. Blobel, and M. P. Rout. 1996. Kap104p: a karyopherin involved in the nuclear transport of messenger RNA binding proteins. Science 274:624-627. Akey, C. W. and M. Raderrnacher. 1993. Architecture of the Xenopus nuclear pore complex revealed by three dimensional cryo-electron microscopy. J. Cell Biol. 122:1-19. Alzhanova-Ericsson, A., X. Sun, N. Visa, E. Kiseleva, T. Wurtz, and B. Daneholt. 1996. A protein of the SR family of splicing factors bind extensively to exonic Balbiani ring pre-mRN A and accompanies the RNA fiom the gene to the nuclear pore. Genes & Devel. 10:2881-2893. Baeuerle, P. A., and D. Baltimore. 1996. NF-sz ten years after. Cell 87:13-20. ll3 Barondes, S. H. 1997. Galectins: A personal overview. Trends in Glycoscience and Glycotechnology 9: 1-7. Barondes, S. H., D. N. W. Cooper, M. A. Gitt, and H. Leffler. 1994. Galectins. J. Biol. Chem. 269:20807-20810. Bastos, R., L. R. de Pouplana, M. Enarson, K. Bodoor, and B. Burke. 1997. Nup84, a novel nucleoporin that is associated with CAN/Nup214 on the cytoplasmic face of the nuclear pore complex. J. Cell Biol. 137:989-1000. Bastos, R., A. Lin, M. Enarson, and B. Burke. 1996. Targeting and function in mRNA export of nuclear pore complex protein Nup153. J. Cell Biol. 134:1141-1156. Bataille, N., T. Helser, and H. M. Fried. 1990. Cytoplasmic transport of ribosomal subunits microinjected into the Xenopus laevis oocyte nucleus: a generalized, facilitated process. J. Cell Biol. 111:1571-1582. Belanger, K. D., M. A. Kenna, S. Wei, and L. I. Davis. 1994. Genetic and physical interactions between Srplp and nuclear pore complex proteins Nuplp and Nup2p. J. Cell Biol. 126:619-630. Bennett, M., S. Michaud, J. Kingston, and R. Reed. 1992. Protein components specifically associated with prespliceosome and spliceosome complexes. Genes & Devel. 6: 1986-2000. Bennett, M. and R. Reed. 1993. Correspondence between a mammalian spliceosome component and an essential yeast splicing factor. Science 262: 105-108. Berger, S. L. and C. S. Birkenmeier. 1979. Inhibition of intractable nucleases with ribonucleoside-vanadyl complexes: isolation of messenger ribonucleic acid from resting lymphocytes. Biochemistry 18:5 143-5 149. Bischoff, F. R., H. Krebber, T. Kempf, I. Hermes, and H. Ponstingl. 1995. Human RanGTPase-activation protein RanGAPl is a homologue of yeast Rnalp involved in mRNA processing and transport. Proc. Natl. Acad. Sci. USA 92:1749-1753. Bischoff, F. R. and H. Ponstingl. 1991. Mitotic regulator protein RCCl is complexed with a nuclear Ras-related polypeptide. Proc. Natl. Acad. Sci. USA 88:10830-10834. 114 Boer, J. M., J. M. A. Van Deursen, H. J. Croes, J. A. M. Fransen, and G. C. Grosveld. 1997. The nucleoporin CAN/Nup214 binds to both the cytoplasmic and the nucleoplasmic sides of the nuclear pore complex in overexpressing cells. Exp. Cell Res. 232:182-185. Bogerd, A. M., J. A. Hoffman, D. C. .Amberg, G. R. Fink, and L. 1. Davis. 1994. nupl mutants exhibit pleiotropic defects in nuclear pore complex function. J. Cell Biol. 127:319—332. Bogerd, H. P., R. A. Fridell, R. E. Benson, J. Hua, and B. R. Cullen. 1996. Protein sequence requirements for function of the human T-cell leukemia virus type 1 Rex nuclear export signal delineated by a novel in vivo randomization-selection assay. Mal. Cel. Biol. 16:4207-4214. Bogerd, H. P., R. A. Fridell, S. Madore, and B. R. Cullen. 1995. Identification of a novel cellular cofactor for the Rev/Rex class of retroviral regulatory proteins. Cell 82:485-494. Bonifaci, N., J. Moroianu, A. Radu, and G. Blobel. 1997. Karyopherin b2 mediates nuclear import of a mRN A binding protein. Proc. Natl. Acad. Sci. USA 94:5055-5060. Borer, R. A., C. F. Lehner, h. M. Eppenberger, and E. A. Nigg. 1989. Major nucleolar proteins shuttle between nucleus and cytoplasm. Cell 56:379-390. Bradford, M. M. 1976. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72:248-254. Buss, F. and M. Stewart. 1995. Macromolecular interactions in the nucleoporin p62 complex of rat nucler pores: binding of nucleoporin p54 to the rod domain of p62. .1. Cell Biol. 128:251-261. Byrd, D. A., D. J. Sweet, N. Pante, K. N. Konstantinov, T. Guan, A. C. S. Saphire, P. J. Mitchell, C. Cooper,S., U. Aebi, and L. Gerace. 1994. Tpr, a large coiled-coil protein whose amino-terminus is involved in activation of oncogenic kinases, is localizaed to the cytoplasmic surface of the nuclear pore complex. J. Cell Biol. 127: 1515-1526. Caceres, J. F., S. Stamm, D. M. Helfrnan, and A. R. Krainer. 1994. Regulation of alternative splicing in vivo by overexpression of antagonistic splicing factors. Science 265:1706-1709. 115 Chandran, U. R. and D. B. DeFranco. 1992. Intemuclear migration of chicken progesterone receptor, but not simian virus-40 large tumor antigen, in transient heterokaryons. Mol. Endocrinol. 6:837-844. Cherayil, B. J., S. J. Weiner, and S. Pillai. 1989. The Mac-2 antigen is a galactose- specific lectin that binds IgE. .1. Exp. Med. 170:1959-1972. Cho, M. and R. D. Cummings. 1995. Galectin-1, a b-galactoside-binding lectin in Chinese hamster ovary cells: 1. Physical and chemical characterization. J. Biol. Chem. 270:5198-5206. Cleves, A. E., D. N. W. Cooper, S. H. Barondes, and R. B. Kelly. 1996. A new pathway for protein export in Saccharomyces cerevisiae. J. Cell Biol. 133:1017-1026. Cooper, D. N., S. M. Massa, and S. H. Barondes. 1991. Endogenous muscle lectin inhibits myoblast adhesion to larninin. J. Cell Biol. 115: 1437-1438. Cooper, D. N. W. and S. H. Barondes. 1990. Evidence for export of a muscle lectin from cytosol to extracellular matrix and for a novel secretory mechanism. J. Cell Biol. 110:1681-1691. Cooper, D. N. W., B. P. Boulianne, S. Charlton, E. M. Farrell, A. Sucher, and B. C. Lu. 1997. Fungal galectins, sequence and specificity of two isolectins from Coprinus cinereus. J. Biol. Chem. 272:1514-1521. Cowles, E. A., N. Agrwal, R. L. Anderson, and J. L. Wang. 1990. Carbohydrate-binding protein 35. Isoelectric points of the polypeptide and a phosphorylated derivative. J. Biol. Chem. 265:17706-17712. Cowles, E. A., I. K. Moutsatsos, J. L. Wang, and R. L. Anderson. 1989. Expression of carbohydrate binding protein 35 in human fibroblasts: comparisons between cells with different proliferative capacities. Exp. Gerontol. 24:577-585. Dagher, S., J. L. Wang, and R. J. Patterson. 1995. Identification of galectin-3 as a factor in pre-mRNA splicing. Proc. Natl. Acad. Sci. USA 92:1213-1217. Daneholt, B. 1997. A look at messenger RNP moving through the nuclear pore. Cell 88:585-588. 116 Dargemont, C. and L. C. Ktihn. 1992. Export of mRNA from microinjected nuclei of Xenopus laevis oocyte. J. Cell Biol. 118:1-9. Davis, L. I. 1995. The nuclear pore complex. Annu. Rev. Biochem. 64:865-896. Davis, L. I. and G. Blobel. 1987. Nuclear pore complex contains a family of glycoproteins that includes p62: Glycosylation through a previously unidentified cellular pathway. Proc. Natl. Acad. Sci. USA 84:7552-7556. Dreyfuss, G., M. J. Matunis, S. Pii‘lol-Roma, and C. G. Burd. 1993. hnRNP proteins and the biogenesis of mRNA. Annu. Rev. Biochem. 62:289-321. Emig, S., D. Schmalz, M. Shakibaei, and K. Buchner. 1995. The nuclear pore complex protein p62 is one of several sialic acid-containing proteins of the nuclear envelope. J. Biol. Chem. 270:13787-13792. Fantozzi, D. A., A. T. Harootunian, W. Wen, S. S. Taylor, J. R. Ferarnisco, R. Y. Tsien, and J. L. Meinkoth. 1994. Therrnostable inhibitor of cAMP-dependent protein kinase enhances the rate of export of the kinase catalytic subunit from the nucleus. J. Biol. Chem. 269:2676-2686. Finlay, D. R., D. D. Newmeyer, T. M. Price, and D. J. Forbes. 1987. Inhibition of in vitro nuclear transport by a lectin that binds to nuclear pores. J. Cell Biol. 104:189-200. Fischer, U., J. Huber, W. C. Boelens, I. W. Mattaj, and R. Lithrrnann. 1995. The HIV-l Rev activation domain is a nuclear export signal that accesses an export pathway used by specific cellular RNAS. Cell 82:475-483. F lach, J., M. Bossie, J. Vogel, A. Corbett, T. Jinks, D. A. Willins, and P. A. Silver. 1994. A yeast RNA-binding protein shuttles between the nucleus and the cytoplasm. Mol. Cell Biol. 14:8399-8407. Fomerod, M., J. van Deursen, S. van Ball, A. Reynolds, .D. Davis, K. G. Murti, J. Fransen, and G. Grosveld. 1997. The human homologue of yeast CRMl is in a dynamic subcomplex with CAN/Nup214 and a novel nuclear pore component Nup88. EMBO J. 16:807-816. 117 Fridell, R. A., R. Truant, L. Thorne, R. E. Benson, and B. R. Cullen. 1997. Nuclear import of hnRNP A1 is mediated by a novel cellular cofactor related to karyopherin-b. J. Cell Sci. 110:1325-1331. Fritz, C. C., M. L. Zapp, and M. R. Green. 1995. A human nucleoporin-like protein that specifically interacts with HIV Rev. Nature 376:530-533. Fukuda, M., I. Gotoh, Y. Goto, and E. Nishida. 1996. Cytoplasmic localization of motigen-activated protein kinase kinase directed by its NHz-terminal, leucine-rich short amino acid sequence, which acts as a nuclear export signal. J. Biol. Chem. 271 :20024- 20028. Gerace, L. 1995. Nuclear export signals and the fast track to the cytoplasm. Cell 82:341- 344. Gerace, L., Y. Ottaviano, and C. Kondor-Koch. 1982. Identification of a major polypeptide Of the nuclear pore complex. J. Cell Biol. 95:826-837. Gitt, M.A., S. M. Massa, H. Leffler; and S. H. Barondes. 1992. Isolation and expression of a gene encoding L-14-II, a new human soluble lactose-binding lectin. J. Biol. Chem 267:10601-10606 Gbrlich, D., R. Kraft, S. Kostka, F. Vogel, E. Hartrnann, R. A. Laskey, I. W. Mattaj, and E. Izaurralde. 1996. Importin provides a link between nuclear import and U snRNA export. Cell 84:21-32. GOrlich, D. and I. W. Mattaj. 1996. Nucleocytoplasmic transport. Science 271:1513-1518. GOrlich, D., S. Prehn, R. A. Laskey, and E. Hartrnann. 1994. Isolation of a protein that is essential for the first step of nuclear protein import. Cell 79:767-778. Greber, U. F. ., A. Senior, and L. Gerace. 1990. A major glycoprotein of the nuclear pore complex is a membrane-spanning polypeptide with a large lumenal domain and a small cytoplasnric tail. EMBO J. 9:1495-1502. Guiochon-Mantel, A., P. Lescop, S. Christin-Maitre, H. Loosfelt, M. Perrot-Applanat, and E. Milgrom. 1991. Nucleocytoplasmic shuttling of the progesterone receptor. EMBO J. 10:3851-3859. 118 Hallberg, E., R. W. Wozniak, and G. Blobel. 1993. An integral membrane protein of the pore membrane domain of the nuclear envelope contains a nucleoporin-like region. J. Cell Biol. 122:513-522. Hamann, K. K., E. A. Cowles, J. L. Wang, and R. L. Anderson. 1991. Expression of carbohydrate binding protein 35 in human fibroblasts: Variations in the levels of mRNA, protein, and isoelectric species as a function of replicative competence. Exp. Cell Res. 196:82-91. Hamm, J. and I. W. Mattaj. 1990. Monomethylated cap structures facilitate RNA export form the nucleus. Cell 63:109-118. Hanada, M., C. Aimé-Sempé, T. Sato, and J. C. Reed. 1995. Structure-function analysis of Bel-2 protein. Identification of conserved domains important for homodimerization with Bcl-2 and heterodirnerization with Bax. J. Biol. Chem. 270:11962-11969. Hart, G. W., G. D. Holt, and R. S. Haltiwanger. 1988. Nuclear and cytoplasmic glycosylation: novel saccharide linkages in unexpected places. T IBS 13:3 80-3 84. Hirabayashi, J ., M. Satoh, and K.-i. Kasai. 1992. Evidence that Caenorhabditis elegans 32-kDa b-galactoside-binding protein is homologous to vertebrate b—galactoside-binding lectins. J. Biol. Chem. 267:15485-15490. Ho, M. K. and T. A. Springer. 1982. Mac-2, a novel 32,000 Mr mouse macrophage subpopulation-specific antigen defined by monoclonal antibodies. J. Immunol. 128: 1221- 1228. Hong, W., M. Bennett, Y. Xiao, R. F. Kramer, C. Wang, and R. Reed. 1997. Association of U2 snRNP with the spliceosomal complex B. Nucleic Acids Res 25:354-361. Hopper, A. K., H. M. Traglia, and R. W. Dunst. 1990. The yeast RNA] gene product necessary for RNA processing is located in the cytosol and apparently excluded from the nucleus. J. Cell Biol. 111:309-321. Hubert, M., S. Y. Wang, J. L. Wang, A. P. Seve, and J. Hubert. 1995. Intranuclear distribution of galectin-3 in mouse 3T3 fibroblasts: Comparative analyses by immunofluorescence and immunoelectron microscopy. Exp. Cell Res. 220:397-406. 119 Huflejt, M. E., C. W. Turck, R. Lindstedt, S. H. Barondes, and H. Leffler. 1993. L-29, a soluble lactose-binding lectin, is phosphorylated on serine 6 and serine 12 in vivo and by casein kinase 1. J. Biol. Chem. 268:26712-26718. Imamoto, N., T. Tachibana, M. Matsubase, and Y. Yoneda. 1995. A karyophilic protein forms a stable complex with cytoplasmic components prior to nuclear pore binding. J. Biol. Chem. 270:8559-8565. Iovine, M. K. and S. R. Wente. 1997. A nuclear export signal in Kap95p is required for both recycling the import factor and interactions with the nucleoporin GLFG repeat regions ofNup116p and Nup100p. J. Cell Biol. 137:797-811. Izaurralde, E., A. Jarmolowski, C. Beisel, I. W. Mattaj, G. Dreyfuss, and U. Fischer. 1997. A role for the M9 transport signal of hnRNP A1 in mRNA nuclear export. J. Cell Biol. 137:27-35. Izaurralde, E., J. Lewis, C. Garnberi, A. Jarmolowski, C. McGuigan, and I. W. Mattaj. 1995. A cap-binding protein complex mediating U snRN A export. Nature 376:709-712. Izaurralde, E., J. Lewis, C. McGuigan, M. Jankowska, E. Darzynkiewicz, and l. W. Mattaj. 1994. A nucler cap bining protein complex involved in pre-mRNA splicing. Cell 78:657-668. Izaurralde, E. and I. W. Mattaj. 1995. RNA export. Cell 81:153-159. Izaurralde, E., J. Stepinski, E. Darzynkiewicz, and I. W. Mattaj. 1992. A cap binding protein that may mediate nuclear export of RNA polymerase II-transcribed RNAS. J. Cell Biol. 118:1287-1295. Jarmolowski, A., W. C. Boelens, E. Izaurralde, and I. W. Mattaj. 1994. Nuclear export of different classes of RNA is mediated by specific factors. J. Cell Biol. 124:627-635. Jia, S. and J. L. Wang. 1988. Carbohysrate binding protein 35. Complementary DNA sequence reveals homology with proteins of the heterogeneous nuclear RNP. J. Biol. Chem. 263:6009-6011. 120 Jiang, L.-W. and M. Schindler. 1987. Fluorescence photobleaching analysis of nuclear transport: dynamic evidence for auxiliary channels in detergent-treated nuclei. Biochemistry 26: 1546-1551 . Kalirrich, J. F. and M. G. Douglas. 1989. In vitro translocation through the yeast nuclear envelope: signal-dependent transport requires ATP and calcium. J. Biol. Chem. 264:17979—17989. Kanigsberg, A., J. Seetharaman, H. Leffler, S. H. Barondes, and J. M. Rini. 1997. Submitted Kasai, K.-i. and J. Hirabayashi. 1996. Galectins: A family of animal lectins that decipher glycocodes. Journal of Biochemistry 119:1-8. Kayden, C. S., J. Seetharaman, H. Leffler, S. H. Barondes, and J. M. Rini.1997. Submitted Kim, F. J., A. A. Beeche, J. J. Hunter, D. J. Chin, and T. J. Hope. 1996. Characterization of the nuclear export signal of human T-cell lymphotropic virus type 1 Rex reveals that nuclear export is mediated by position-variable hydrophobic interaction. Mal. Cel. Biol. 16:5147-5155. Koepp, D. M. and P. A. Silver. 1996. A GTPase controlling nuclear trafficking: running the right way or walking RANdomly? Cell 87:1-4. Kraemer, D., R. W. Wozniak, G. Blobel, and A. Radu. 1994. The human CAN protein, a putative oncogene product associated with myeloid leukemogeneisis, is a nuclear pore complex protein that faces the cytoplasm. Proc. Natl. Acad. Sci. USA 91 :1519-1523. Kutay, U., E. Izaurralde, F. R. Bischoff, I. W. Mattaj, and D. GOrlich. 1997. Dominant- negative mutants of importin-b block multiple pathways of import and export through the nuclear pore complex. EMBO J. 16:1153-1163. Laing, J. G. and J. L. Wang. 1988. Identification of carbohydrate binding protein 35 in heterogeneous nuclear ribonucleoprotein complex. Biochemistry 27:5329—5334. 121 Lee, M. S., M. Henry, and P. A. Silver. 1996. A protein that shuttles between the nucleus and the cytoplasm is an important mediator of RNA export. Genes & Devel. 10:1233- 1246. Lefller, H. 1997. Introduction to galectins. Trends in Glycoscience and Glycotechnology 9:9—19. Lewis, J. D., D. GOrlich, and I. W. Mattaj. 1996. A yeast cap binding protein complex (yCBC) acts at an early step in pre-mRNA splicing. Nucleic Acids Res 24:3332-3336. Li, J., G. Elberg, D. C. Crans, and Y. Shechter. 1996. Evidence for the distinct vanadyl(+4)-dependent activating system for manifesting insulin-like effects. Biochemistry 35: 8314-8318 Liao, D.-I., G. Kapadia, H. Ahmed, G. R. Vasta, and O. Herzberg. 1994. Structure of S- lectin, a developmentally regulated vertebrate B-galactoside-binding protein. Proc. Natl. Acad. Sci. USA 91:1428-1432. Liao, X. C., H. V. Colot, Y. Wang, and M. Rosbash. 1992. Requirements for U2 snRNP addition to yeast pre-mRNA. Nucleic Acids Res 20:4237-4235. Lindstedt, R., G. Apdaca, S. H. Barondes, K. E. Mostov, and H. Leffler. 1993. Apical secretion of a cytosolic protein by Madin-Darby canine kidney cells. J. Biol. Chem. 268:11750-11757. Lobsanov, Y. D., M. A. Gitt, H. Leffler, S. H. Barondes, and J. M. Rini.1993. X-ray crystal structure of the human dimeric S-lac lectin, L-14-II, in complex with lactose at 2.9-A resolution. J. Biol. Chem. 268:27034-27038. Lobsanov, Y. D. and J. M. Rini. 1997. Galectin structure. Trends in Glycoscience and Glycotechnology 9: 145-154. Lotz, M. M., C. W. Andrews,Jr., C. A. Korzelius, E. C. Lee, G. D. Steele,Jr., A. Clarke, and A. M. Mercurio. 1993. Decreased expression of Mac-2 (carbohydrate binding protein 35) and loss of its nuclear localization are associated with the neoplastic progression of colon carcinoma. Proc. Natl. Acad. Sci. U. S. A. 90:3466-3470. 122 Macara, I. G., K. Kustin, and J.C. Cantley Jr. 1980. Glutathione reduces cytoplasmic vanadate. Mechanism and physiological implications. Biochim. Biophys. Acta 629:95-106 Mandel], R. B. and C. M. Feldherr. 1990. Identification of two HSP70-related Xenopus oocyte proteins that are capable of recycling across the nuclear envelope. J. Cell Biol. 111:1775-1783. Marshallsay, C. and R. Liihrmann. 1994. In vitro nuclear import of snRNPs: cytosolic factors mediate m3G-cap dependence of U1 and U2 snRNP transport. EMBO J. 13:222- 231. Mayeda, A. and A. R. Krainer. 1992. Regulation of alternative pre-mRNA splicing by hnRNP Al and splicing factor SF2. Cell 68:365-375. Melchior, F., T. L. Guan, N. Yokoyarna, T. Nishirnoto, and L. Gerace. 1995. GTP hydrolysis by Ran occurs at the nuclear pore complex in an early step of protein import. J. Cell Biol. 131:571-581. Melchior, F., B. M. Paschal, J. Evans, and L. Gerace. 1993. Inhibition of nuclear protein import by nonhybrolyzable analogues of GTP and identification Of the small GTPase Ran/TC4 as an essential transport factor. J. Cell Biol. 123:1649-1659. Meyer, B. E. and M. H. Malirn. 1994. The HIV-1 Rev trans-activator shuttles between the nucleus and the cytoplasm. Genes-Dev. 8: 1538-1 547. Michaud, S. and R. Reed. 1991. An ATP-independent complex commits pre-mRNA to the mammalian spliceosome assembly pathway. Genes & Devel. 5:2534-2546. Michael, W. M., M. Y. Choi, and G. Dreyfuss. 1995. A nuclear export signal in hnRNP A1: A signal-mediated, temperature-dependent nuclear protein export pathway. Cell 83:415-422. Michael, W. M., P. S. Eder, and G. Dreyfuss. 1997. The K nuclear shuffling domain: a novel signal for nuclear import and nuclear export in the hnRNP K protein. EMBO J. 16:3587-3598. Misaki, Y., G. J. Pruijn, A. W. van der Kemp, and W. J. van Venrooij. 1994. The 56K autoantigen is identical to human annexin XI. J. Biol. Chem. 269:4240-4246. 123 Mizutani, A., N. Watanabe, T. Kitao, H. Tokunritsu, and H. Hidaka. 1995. The long amino-terminal tail domain of annexin XI is necessary for its nuclear localization. Arch. Biochem. Biophys. 318: 1 57-165. Moore, M. J., C. C. Query, and P. A. Sharp. 1993. Splicing of precursors of mRNA by the spliceosome. In The RNA World. R. F. Gesteland and J. F. Atkins, editors. Cold Spring Harbor Laboratory Press, New York. 303-357. Moore, M. S. 1996. Protein translocation: Nuclear export - out of the dark. Current Biology 6: 137-140. Moore, M. S. and G. Blobel. 1993. The GTP-binding protein Ran/T C4 is required for protein import into the nucleus. Nature 365:661-663. Moore, M. S. and G. Blobel. 1994. Purification of a Ran-interacting protein that is required for protein import into the nucleus. Proc. Natl. Acad. Sci. USA 91 : 10212-10216. Moroianu, J. and G. Blobel. 1995. Protein export from the nucleus requires the GTPase Ran and GTP hydrolysis. Proc. Natl. Acad Sci. USA 92:4318-4322. Moroianu, J ., G. Blobel, and A. Radu. 1995. Previously identified protein of uncertain function is karyopherin a and together with karyopherin b docks import substrate at nuclear pore complexes. Proc. Natl. Acad. Sci. USA 92:2008-2011. Moroianu, J ., G. Blobel, and A. Radu. 1996. The binding site of karyopherin a for karyopherin b overlaps with a nuclear localization sequence. Proc. Natl. Acad. Sci. USA 93:6572-6576. Moroianu, J ., G. Blobel, and A. Radu. 1996. Nuclear protein import: Ran-GTP dissociate the karyopherin ab heterodimer by displaing a from and overlapping binding site on b. Proc. Natl. Acad. Sci. USA 93:7059-7062. Moroianu, J., M. Hijikata, G. Blobel, and A. Radu. 1995. Mammalian karyopherin a,b and ab heterodimers: al or a2 sububit binds nuclear localization signal and b subunit interacts with peptide repeat-containing nucleoporins. Proc. Natl. Acad Sci. USA 92:6532-6536. 124 Mostov, K., G. Apodaca, B. Aroeti, and C. Okarnoto. 1992. Plasma membrane protein sorting in polarized epithelial cells. J. Cell Biol. 116:577-583. Moutsatsos, I. K., J. M. Davis, and J. L. Wang. 1986. Endogenous lectins from cultured cells: Subcellular localization of carbohydrate-binding protein 35 in 3T3 fibroblasts. J. Cell Biol. 102:477-483. ' Moutsatsos, I. K., M. Wade, M. Schindler, and J. L. Wang. 1987. Endogenous lectins from cultured cells: nuclear localization of carbohydrate-binding protein 35 in proliferating cells. Proc. Natl. Acad. Sci. USA 84:6452-6456. Muesch, A., E. Hartrnann, K. Rohde, A. Rubartelli, R. Sitia, and T. A. Rapoport. 1990. A novel pathway for secretory proteins? TIBS 15:86-88. Nehrbass, U. and G. Blobel. 1996. Role of the nuclear transport factor p10 in nuclear import. Science 272: 120-122. Neuman de Vegvar, H. E. and J. E. Dahlberg. 1990. Nucleocytoplasmic transport and processing of small nuclear RNA precursors. Mol. Cel. Biol. 10:2265-3375. Newmeyer, D. D. and D. J. Forbes. 1988. Nuclear import can be separated into distinct steps in vitro: nuclear pore binding and translocation. Cell 52:641-653. Nigg, E. A. 1997. Nucleocytoplasmic transport: Signals, mechanisms and regulation. Nature 386:779-787. Numinskaya, M. and T. F. Linsenmayer. 1996. Identification of characterization of up- regulated genes during Chondrocyte hypertrophy. Developmental Dynamics 206:260-271. O'Farrell, P. Z., H. M. Goodman, and P. H. O’Farrell. 1977. High resolution two- dimensional electrophoresis of basic as well as acidic proteins. Cell 1977 12:1133-1141 Pante, N., R. Bastos, I. McMorrow, B. Burke, and U. Aebi. 1994. Interactions and three dimensional localization of a group of nuclear pore complex proteins. J. Cell Biol. 126:603-617. 125 Paschal, B. M. and L. Gerace. 1995. Identification of NTF2, a cytosolic factor for nuclear import that interacts with nuclear pore complex protein p62. J. Cell Biol. 129:925-93 7. Patterson, R. J ., S. F. Dagher, A. Vyakamam, and J. L. Wang. 1997. Nuclear galectins: Functionally redundant components in processing of pre-mRN A. Trends in Glycoscience and Glycotechnology 9:77-85. Perillo, N. L., K. E. Pace, J. J. Seilharner, and L. G. Batun. 1995. Apoptosis of T cells mediated by galectin-l. Nature 378:736-739. Perillo, N. L., C. H. Uittenbogaart, J. T. Nguyen, and L. G. Baum. 1997. Galectin-1, an endogenous lectin produced by thymic epithelial cells, induces apoptosis of human thymocytes. J. Exp. Med. 185:1851-1858. Peters, R 1986. Fluorescence rnicrophotolysis to measure nucleocytoplasrrric transport and intracellular mobility. Biochim. Biophys. Acta 864:305-359. Pifiol-Roma, S., Y. D. Choi, M. J. Matunis, and G. Dreyfuss. 1988. Immunopurification of heterogeneous nuclear ribonucleoprotein particles reveals an assortment of RNA- binding proteins. Genes-Dev. 2:215-227. PiflOl-Roma, S. and G. Dreyfuss. 1993. Shuttling of pro-mRNA binding proteins between nucleus and cytoplasm. Nature 355:730-732. Piotrowicz, R. S., J. L. Martin, W. H. Dilhnan, and E. G. Levin. 1997. The 27-kDa heat shock protein facilitates basic fibroblast growth factor release from endotlrelial cells. J. Biol. Chem. 272:7042-7047. Pollard, V. W., W. M. Michael, S. NAkielny, M. C. Sionri, F. Wang, and G. Dreyfuss. 1996. A novel receptor-mediated nuclear import pathway. Cell 86:985-994. Powers, M. A., D. J. Forbes, J. E. Dalrlberg, and E. Lund. 1997. The vertebrate GLFG nucleoporin, Nup98, is an essential component of multiple RNA export pathway. J. Cell Biol. 136:241-250. Powers, M. A., C. Macaulay, F. R. Masiarz, and D. J. Forbes. 1995. Reconstituted nuclei depleted of a vertebrate GLF G nuclear pore protein, p97, import but are defective in nuclear growth and replication. J. Cell Biol. 128:721-736. 126 Puch, A. C. and B. Key. 1995. Identification of cells expressing galectin-l, a galactose- binding receptor, in the rat Olfactory system. The Journal of Comparative Neurology 357:513-523. Puche, A. C., F . Poirier, M. Hair, P. F. Bartlee, and B. Key. 1996. Role of galectin-1 in the developing mouse olfactory system. Developmental Biology 179:274-287. Radu, A., G. Blobel, and R. W. Wozniak. 1993. NuplSS is a novel nuclear pore complex protein that contains neither repititive sequence motifs nore reacts with WGA. J. Cell Biol. 121:1-9. Radu, A., G. Blobel, and R. W. Wozniak. 1994. Nup107 is a novel nuclear pore complex protein that contains a leucine zipper. J. Biol. Chem. 269:17600-17605. Radu, A., M. S. Moore, and G. Blobel. 1995. The peptide repeat domain of nucleoporin Nup98 functions as a docking site in transport across the nuclear pore complex. Cell 81:215-222. Rexach, M. and G. Blobel. 1995. Protein import into nuclei: association and dissociation reactions involving transport substrate, transport factors, and nucleoporins. Cell 83:683- 692. Richards, S. A., K. L. Carey, and I. G. Macara. 1997. Requirement of guanosine triphosphate-bound Ran for signal-mediated nuclear protein export. Science 276: 1842- 1844. Richardson, W. D., A. D. Mills, S. M. Dilworth, R. A. Laskey, and C. Dingwall. 1988. Nuclear protein migration involves two steps: rapid binding at the nuclear envelope followed by slower translocation through nuclear pores. Cell 52:655-664. Roff, C. F ., P. R. Rosevear, J. L. Wang, and R. Barker. 1983. Identification of carbohydrate-binding proteins from mouse and human fibroblasts. Biochem. J. 211:625- 629. Rothmarr, J. E. and L. Orci. 1992. Molecular dissection of the secretory pathway. Nature 355:409-415. 127 Sachs, A. B., P. Samow, and M. W. Hentze. 1997. Starting at the beginning, middle, and end: translation initiation in eukaryocytes. Cell 89:831-83 8. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Molecular cloning. Cold Spring Harbor Laboratory Press, New York. Siders, W. M. and S. B. Mizel. 1995. Interleukin-1b secretion. J. Biol. Chem. 270:16258- 16264. Sirnons, K. and A. Wandinger-Ness. 1990. Polarized sorting in epithelia. Cell 62:207- 210. Sionri, H. and G. Dreyfuss. 1995. A nuclear localization domain in the hnRNP A1 protein. J. Cell Biol. 129:551-560. Smalheiser, N. R. 1996. Proteins in unexpected locations. Mol. Biol. Cell 7:1003-1014. Snow, C. M., A. Senior, and L. Gerace. 1987. Monoclonal antibodies identify a group Of nuclear pore complex glycoproteins. J. Cell Biol. 104:1143-1156. Spector, D. L. 1996. Nuclear organization and gene expression. Exp. Cell Res. 229:189- 197. Starr, C. M., M. D'Onofrio, M. K. Park, and J. A. Hanover. 1990. Primary sequence and heterologous expression of nuclear pore glycoprotein p62. J. Cell Biol. 1 10: 1861-1871. Sukegawa, J. and G. Blobel. 1993. A nuclear pore complex protein that contains zinc fingers and faces the nucleoplasm. Cell 72:29-38. Tarantini, F., S. Gamble, A. Jackson, and T. Maciag. 1995. The cysteine residue responsible for the release of fibroblast growth factor-1 resides in a domain independent of the domain for phsophatidylserine binding. .1. Biol. Chem. 270:29039-29042. Tokunritsu, H., A. Mizutarri, H. Minarni, R. Kobayashi, and H. Hidaka. 1992. A calcyclin-associated protein is a newly identified member of the Cazi/phospholipid- binding proteins, annexin family. J. Biol. Chem. 267:8919-8924. 128 Valcarcel, J. and M. R. Green. 1996. The SR protein family: pleiotropic functions in pre- mRNA splicing. TIBS 21:296-301. Visa, N., A. Alzhanova-Ericsson, X. Sun, E. Kiseleva, B. BjOrkroth, T. Wurtz, and B. Daneholt. 1996. A pre-mRNA-binding protein accompanies the RNA from the gene through the nuclear pores and into polysomes. Cell 84:253-264. Visa, N., E. Izaurralde, J. Ferreira, B. Daneholt, and I. W. Mattaj. 1996. A nuclear cap- binding complex binds Balbiani ring pre-mRNA cotranscriptionally and accompanies the ribonucleoprotein particle during nuclear export. J. Cell Biol. 133:5-14. Vyakamam, A. 1996. Galectin-l in the cell nucleus: Evidence for a role in pre-mRNA . splicing. Ph. D. Dissertation. Vyakamam, A., S. F. Dagher, J. L. Wang, and R. J. Patterson. 1997. Evidence for a role for galectin-l in pre-mRNA splicing. Mal. Cel. Biol. 17:4730-4737. Wada, J., K. Ota, A. Kumar, E. I. Wallner, and Y. S. Kanwar. 1997. Developmental regulation, expression, and apoptotic potential of galectin-9, a B-galactoside binding lectin. J. Clin Invest. 99:2452-2461. Wang, J. L., Anandita, S.-Y. Wang, and N. Agrwal. 1993. Animal lectins: Carbohydrate binding protein 35: Production of the recombinant polypeptide and its individual domains. In Lectins: Biology, biochemistry, clinical biochemistry. J. Basu, M. Kundu, and P. Chakrabarti, editors. Wiley Eastern Limited, New Delhi. 89-99. Wang, J. L., J. G. Laing, and R. L. Anderson. 1991. Lectins in the cell nucleus. Glycobiology. 1:243-252. Wang, L., H. Inohara, K. J. Pienta, and A. Raz. 1995b. Galectin-3 is a nuclear matrix protein which binds RNA. Biochem. Biophys. Res. Commun. 217:292-303. Wang, S.-Y., P. G. Voss, R. J. Patterson, and J. L. Wang. 1995a. Studies on the cell surface versus nuclear localization of galectin-3. Antibody, Immunoconjugates, and Radiopharmaceuticals 8 :31 1-3 24. 129 Weighardt, F., G. Biarnonti, and S. Riva. 1996. The roles of heterogeneous nuclear ribonucleoproteins (hnRNP) in RNA metabolism. BioEssays 18:747-756. Weis, K., C. Dingwall, and A. I. Lamond. 1996. Characterization of the nuclear protein import mechanism using Ran mutants with altered nucleotide binding specificities. EMBOJ. 15:7120—7128. Weller, P. F., D. S. Bach, and K. F. Austen. 1984. Biochemical characterization of hmnan eosinophil Charcot-Leyden crystal protein (lysophospholipase). J. Biol. Chem. 259:15100-15105. Wen, W., J. L. Meinkotlr, R. Y. Tsien, and S. S. Taylor. 1995. Identification of a signal for rapid export of proteins from the nucleus. Cell 82:463-473. Wilken, N., U. Kossner, J.-L. Senécal, U. Scheer, and M.-C. Dabauvalle. 1993. Nup180, a novel nuclear pore complex protein localizing to the cytoplasmic ring and associated fibrils. J. Cell Biol. 123:1345-1354. Wolff, B., M. C. Willingharn, and J. A. Hanover. 1988. Nuclear protein import: specificity for transport across the nuclear pore. Exp. Cell Res. 178:3 1 8-3 34. Wozniak, R. W., E. Bartrrik, and G. Blobel. 1989. Primary structure analysis of an integral membrane glycoprotein of the nuclear pore. J. Cell Biol. 108:2083-2092. Wozniak, R. W. and G. Blobel. 1992. The single transmembrane segment of ngIO is sufficient for sorting to the pore membrane domain of the nuclear envelope. J. Cell Biol. 119:1441-1449. Wu, J., M. J. Matunis, D. Kraemer, G. Blobel, and E. Coutavas. 1995. Nup358, a cytoplasmc exposed nucleoporin with peptide repeats, Ran-GTP binding sites, zinc fingers, a cyclophilin A homologous domain, and a leucine-rich region. J. Biol. Chem. 270:14209-14213. Wu, J. Y. and T. Maniatis. 1993. Specific interactions between proteins implicated in splice site selection and regulated altemative splicing. Cell 75: 1061-1070. Yang, J., J. Liu, and D. B. DeFranco. 1997. Subnuclear trafficking of glucocorticoid receptors in vitro: Chromatin recycling and nuclear export. J. Cell Biol. 137:523-538. 130 Yang, R.-Y., D. K. Hsu, and F.-T. Liu. 1996. Expression of galectin-3 modulates T-cell growth and apoptosis. Proc. Natl. Acad. Sci. USA 93:6737-6742. Yaseen, N. R. and G. Blobel. 1997. Cloning and characterization of human karyopherin b3. Proc. Natl. Acad. Sci. USA 94:4451-4456. Yin, X.-M., Z. N. Oltval, and S. J. Korsmeyer. 1994. 8H] and BH2 domains of Bcl-2 are required for inhibiton of apoptosis and heterodirnerization with Bax. Nature 369:321-323. Yokoyama, N., N. Hayashi, T. Seki, N. Pante, T. Ohba, K. Nishii, K. Kuma, T. Hayashida, T. Miyata, U. Aebi, M. Fukui, and T. Nishirnoto. 1995. A giant nucleopore protein that binds Ran/TC4. Nature 376: 184-1 88. Yoneda, Y., N. Imamato-Sonobebe, Y. Matsuoka, R. Iwarnoto, YiKiho, and T. Uchida. 1988. Antibodies to Asp-Asp-Glu-Asp can inhibit transport of nuclear proteins into the nucleus. Science 242:276-278. Zilhnann, M., M. L. Zapp, and S. M. Berget. 1988. Gel electrophoretic isolation of splicing complexes containing U1 small nuclear ribonucleoprotein particles. Mal. Cel. Biol. 8:814-821. Zolotukhin, A. S. and B. K. Felber. 1997. Mutations in the nuclear export signal of human Ran-binding protein RanBPl block the Rev-mediated posttranscriptional regulation of human immunodeficiency virus type 1. J. Biol. Chem. 272:11356-11360. MICHIGAN srnTE UNIV. LIBRARIES lllllllllllllllllllllllllllllllllllllllllllllllllllllllll 31293017126511